Which finding in a patients history would alert the nurse to assess for signs of hypophosphatemia

Journal Article

Erik A. Imel,

1Departments of Medicine (E.A.I., M.J.E.), Indianapolis, Indiana 46202

2Pediatrics (E.A.I.), Indianapolis, Indiana 46202

Search for other works by this author on:

Michael J. Econs

1Departments of Medicine (E.A.I., M.J.E.), Indianapolis, Indiana 46202

3Medical and Molecular Genetics (M.J.E.), Indiana University School of Medicine, Indianapolis, Indiana 46202

*Address all correspondence and requests for reprints to: Michael J. Econs, M.D., F.A.C.P., F.A.C.E., Division of Endocrinology and Metabolism, Indiana University School of Medicine, 541 North Clinical Drive, CL 459, Indianapolis, Indiana 46202.

Search for other works by this author on:

Accepted:

02 November 2011

  • PDF
  • Split View
    • Article contents
    • Figures & tables
    • Video
    • Audio
    • Supplementary Data
  • Cite

    Cite

    Erik A. Imel, Michael J. Econs, Approach to the Hypophosphatemic Patient, The Journal of Clinical Endocrinology & Metabolism, Volume 97, Issue 3, 1 March 2012, Pages 696–706, //doi.org/10.1210/jc.2011-1319

    Close

    • Email
    • Twitter
    • Facebook
    • More

Close

Navbar Search Filter Microsite Search Term Search

Hypophosphatemia is commonly missed due to nonspecific signs and symptoms, but it causes considerable morbidity and in some cases contributes to mortality. Three primary mechanisms of hypophosphatemia exist: increased renal excretion, decreased intestinal absorption, and shifts from the extracellular to intracellular compartments. Renal hypophosphatemia can be further divided into fibroblast growth factor 23-mediated or non-fibroblast growth factor 23-mediated causes. Proper diagnosis requires a thorough medication history, family history, physical examination, and assessment of renal tubular phosphate handling to identify the cause. During the past decade, our understanding of phosphate metabolism has grown greatly through the study of rare disorders of phosphate homeostasis. Treatment of hypophosphatemia depends on the underlying disorder and requires close biochemical monitoring. This article illustrates an approach to the hypophosphatemic patient and discusses normal phosphate metabolism.

Case

A 65-yr-old man developed right medial knee pain while golfing. A tibial stress fracture was identified. He then developed a contralateral stress fracture and generalized pain and weakness in his legs and back. He had no previous history of fracture or childhood rickets. He has type 2 diabetes, hypercholesterolemia, and hypertension, treated with glipizide, quinapril, rosiglitazone, and atorvastatin. He was taking hydrocodone/acetaminophen, rofecoxib, and tramadol for pain, plus 1000 mg of calcium and 600 units of vitamin D daily. Family history was unremarkable.

On examination, he was hypertensive (147/81 mm Hg), weight was 99.2 kg, and height was 182 cm. He had normal dentition, without intraoral masses. He had a cataract in his left eye. He had no palpable masses in his neck or extremities. Examination of lungs, heart, and abdomen was normal, except for a systolic murmur. He required a walker to ambulate, used his arms to rise from a chair, but could do a sit-up. Laboratory testing revealed calcium of 8.9 mg/dl, creatinine of 0.8 mg/dl, 25-hydroxyvitamin D (25OHD) of 26 ng/ml, 1,25-dihydroxyvitamin D (1,25OHD) of 25 pg/ml, alkaline phosphatase that had risen from 66 to 171 U/liter, and serum phosphate ranging from 1.1 to 1.7 mg/dl.

Accreditation and Credit Designation Statements

The Endocrine Society is accredited by the Accreditation Council for Continuing Medical Education to provide continuing medical education for physicians. The Endocrine Society has achieved Accreditation with Commendation.

The Endocrine Society designates this Journal-based CME activity for a maximum of 1 AMA PRA Category 1 Credit™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.

Learning Objectives

Upon completion of this educational activity, participants should be able to:

  • •. 

    Explain the basic physiology and pathophysiology of phosphates.

  • •. 

    Recognize the causes and symptoms of hypophosphatemia and select appropriate diagnostic testing.

  • •. 

    Apply strategies to diagnose and manageacute and chronic hypohosphatemia.

Target Audience

This Journal-based CME activity should be of substantial interest to endocrinologists.

Disclosure Policy

Authors, editors, and Endocrine Society staff involved in planning this CME activity are required to disclose to learners any relevant financial relationship(s) that have occurred within the last 12 months with any commercial interest(s) whose products or services are discussed in the CME content. The Endocrine Society has reviewed all disclosures and resolved or managed all identified conflicts of interest, as applicable.

Disclosures for JCEM Editors are found at //www.endo-society.org/journals/Other/faculty_jcem.cfm.

The following individuals reported relevant financial relationships:

Michael J. Econs, M.D., is a consultant to and receives royalties from, Kyowa Hakko Kirin Pharma, Inc.

Eric Imel, M.D., is an investigator for Kyowa Hakko Kirin Pharma, Inc.

Editor-in-Chief, Leonard Wartofsky, M.D., reported no relevant financial relationships.

Endocrine Society staff associated with the development of content for this activity reported no relevant financial relationships.

Acknowledgement of Commercial Support

This activity is not supported by grants, other funds, or in-kind contributions from commercial supporters.

Privacy and Confidentiality Statement

The Endocrine Society will record learner's personal information as provided on CME evaluations to allow for issuance and tracking of CME certificates. No individual performance data or any other personal information collected from evaluations will be shared with third parties.

Method of Participation

This Journal-based CME activity is available in print and online as full text HTML and as a PDF that can be viewed and/or printed using Adobe Acrobat Reader. To receive CME credit, participants should review the learning objectives and disclosure information; read the article and reflect on its content; then go to //jcem.endojournals.org and find the article, click on CME for Readers, and follow the instructions to access and complete the post-activity test questions and evaluation.

To complete this activity, participants must:

  • •. 

    Have access to a computer with an internet connection.

  • •. 

    Use a major web browser, such as Internet Explorer 7+, Firefox 2+, Safari, Opera, or Google Chrome; in addition, cookies and Javascript must be enabled in the browser's options.

The estimated time to complete this activity, including review of material, is 1 hour. If you have questions about this CME activity, please direct them to .

Activity release date: March 2012

Activity expiration date: March 2013

Background

Phosphate is involved in a variety of processes including acid-base buffering, postreceptor signaling, energy transfer, and information storage and translation in DNA and RNA. The primary repository (85%) of phosphate is in the bone, where calcium and phosphate (in hydroxyapatite) provide skeletal strength and rigidity (1). Outside of bone, most phosphate is intracellular.

Phosphate is abundantly present in many foods. Isolated dietary phosphate deficiency is uncommon, and deficiency usually occurs with generalized malnutrition. Intestinal phosphate absorption is up-regulated by 1,25OHD. In equilibrium, about 300 mg of phosphate moves into and out of bone daily in adults. Phosphate is freely filtered at the glomerulus and then reabsorbed, but proximal tubular reabsorption is inhibited by both PTH and fibroblast growth factor 23 (FGF23).

For adults, hypophosphatemia is generally defined by serum phosphate concentration below 2.5 mg/dl (0.8 millimolar). However, infants have higher normal phosphate ranges than adults, and these values gradually decline throughout childhood and adolescence to adult levels (2, 3). Young children also have higher renal phosphate thresholds than adults (tubular maximum reabsorption of phosphate per unit of glomerular filtrate, TmP/GFR) (2). This is likely an accommodation to an increased need for phosphate to adequately mineralize the growing skeleton because phosphate concentrations within the adult normal range cause rickets in infancy. Many clinical laboratories do not report age-appropriate normal ranges for phosphate, resulting in missed or delayed diagnoses of hypophosphatemia in children.

Clinical Presentation

Symptoms are nonspecific and are more common with severe and acute hypophosphatemia. Many patients with mild hypophosphatemia are asymptomatic, and hypophosphatemia may be an incidental finding. Generalized muscle weakness is the most common symptom of hypophosphatemia, and weakness and fatigue are frequent symptoms with acquired hypophosphatemia. However, weakness is infrequent in patients with congenital forms, such as X-linked hypophosphatemia (XLH). Other neurological symptoms (including paresthesias, dysarthria, altered mental status, seizures, and neuropathy) are reported with severe hypophosphatemia, but these are rare presenting symptoms for hypophosphatemia in general (4–7). Acute severe hypophosphatemia can be life-threatening and is associated with mortality and impaired cardiac and respiratory function among hospitalized patients (8).

Myalgia may accompany weakness in some patients, but severe acute muscle pain may indicate rhabdomyolysis resulting from hypophosphatemia. Hypophosphatemia may also cause intravascular hemolysis. During cell lysis, intracellular phosphate and proteins such as myoglobin are released, which can cause renal damage and hyperphosphatemia, obscuring previous hypophosphatemia.

Patients with chronic hypophosphatemia develop osteomalacia and resultant bone pain. Pain may occur with or without pseudofractures. However, osteomalacia requires time to develop and is not present during acute hypophosphatemia. Growing children with prolonged hypophosphatemia from any etiology develop rachitic features, including genu valgum or varum, frontal bossing, widening of the ends of long bones, and short stature.

A careful history of both prescription and nonprescription medications may identify the cause of hypophosphatemia (Table 1). In addition, family history may reveal a heritable disorder, although some patients with inherited hypophosphatemia may be unaware of their family diagnosis (9).

Table 1.

Differential diagnosis of hypophosphatemia

Increased renal excretionImpaired intestinal absorption or intakeTranscellular shiftsOthersFGF23-mediatedNon-FGF23-mediated
XLH (PHEX)  Hyperparathyroidism  Impaired dietary intake  Refeeding syndrome  Mannitol 
ADHR (FGF23)  HHRH  Phosphate binders  Glucose infusion  Bisphosphonates 
ARHR (DMP1, ENPP1)  Diuretics: acetazolamide, thiazides, loop diuretics  Sevelamer  Insulin infusion   
TIO    Antacids containing calcium, magnesium, aluminum  Salicylate poisoning   
FD  Fanconi syndrome    Hyperventilation   
Linear sebaceous nevus syndrome  Genetic causes: Dent's disease, cystinosis, NaPi2a mutations, others  Alcoholism  Respiratory alkalosis   
Postrenal transplantation hypophosphatemia  Drug induced: toluene, streptozocin, ifosfamide, cisplatin, tetracyclines, aminoglycosides, antiretrovirals (tenofovir, adefovir), and imatinib  Premature infants  Catecholamines   
Iron polymaltose infusions    Malabsorption     
    Vitamin D deficiency     
    Vitamin D metabolism defects     
    1α-hydroxylase deficiency     
    Vitamin D receptor mutation     

Increased renal excretionImpaired intestinal absorption or intakeTranscellular shiftsOthersFGF23-mediatedNon-FGF23-mediated
XLH (PHEX)  Hyperparathyroidism  Impaired dietary intake  Refeeding syndrome  Mannitol 
ADHR (FGF23)  HHRH  Phosphate binders  Glucose infusion  Bisphosphonates 
ARHR (DMP1, ENPP1)  Diuretics: acetazolamide, thiazides, loop diuretics  Sevelamer  Insulin infusion   
TIO    Antacids containing calcium, magnesium, aluminum  Salicylate poisoning   
FD  Fanconi syndrome    Hyperventilation   
Linear sebaceous nevus syndrome  Genetic causes: Dent's disease, cystinosis, NaPi2a mutations, others  Alcoholism  Respiratory alkalosis   
Postrenal transplantation hypophosphatemia  Drug induced: toluene, streptozocin, ifosfamide, cisplatin, tetracyclines, aminoglycosides, antiretrovirals (tenofovir, adefovir), and imatinib  Premature infants  Catecholamines   
Iron polymaltose infusions    Malabsorption     
    Vitamin D deficiency     
    Vitamin D metabolism defects     
    1α-hydroxylase deficiency     
    Vitamin D receptor mutation     

See Refs. 1, 7, 14, 18–21, 23, 26, 27, 30–36, 39, 40, 43–46, 53, 70, 78–81.

Table 1.

Differential diagnosis of hypophosphatemia

Increased renal excretionImpaired intestinal absorption or intakeTranscellular shiftsOthersFGF23-mediatedNon-FGF23-mediated
XLH (PHEX)  Hyperparathyroidism  Impaired dietary intake  Refeeding syndrome  Mannitol 
ADHR (FGF23)  HHRH  Phosphate binders  Glucose infusion  Bisphosphonates 
ARHR (DMP1, ENPP1)  Diuretics: acetazolamide, thiazides, loop diuretics  Sevelamer  Insulin infusion   
TIO    Antacids containing calcium, magnesium, aluminum  Salicylate poisoning   
FD  Fanconi syndrome    Hyperventilation   
Linear sebaceous nevus syndrome  Genetic causes: Dent's disease, cystinosis, NaPi2a mutations, others  Alcoholism  Respiratory alkalosis   
Postrenal transplantation hypophosphatemia  Drug induced: toluene, streptozocin, ifosfamide, cisplatin, tetracyclines, aminoglycosides, antiretrovirals (tenofovir, adefovir), and imatinib  Premature infants  Catecholamines   
Iron polymaltose infusions    Malabsorption     
    Vitamin D deficiency     
    Vitamin D metabolism defects     
    1α-hydroxylase deficiency     
    Vitamin D receptor mutation     

Increased renal excretionImpaired intestinal absorption or intakeTranscellular shiftsOthersFGF23-mediatedNon-FGF23-mediated
XLH (PHEX)  Hyperparathyroidism  Impaired dietary intake  Refeeding syndrome  Mannitol 
ADHR (FGF23)  HHRH  Phosphate binders  Glucose infusion  Bisphosphonates 
ARHR (DMP1, ENPP1)  Diuretics: acetazolamide, thiazides, loop diuretics  Sevelamer  Insulin infusion   
TIO    Antacids containing calcium, magnesium, aluminum  Salicylate poisoning   
FD  Fanconi syndrome    Hyperventilation   
Linear sebaceous nevus syndrome  Genetic causes: Dent's disease, cystinosis, NaPi2a mutations, others  Alcoholism  Respiratory alkalosis   
Postrenal transplantation hypophosphatemia  Drug induced: toluene, streptozocin, ifosfamide, cisplatin, tetracyclines, aminoglycosides, antiretrovirals (tenofovir, adefovir), and imatinib  Premature infants  Catecholamines   
Iron polymaltose infusions    Malabsorption     
    Vitamin D deficiency     
    Vitamin D metabolism defects     
    1α-hydroxylase deficiency     
    Vitamin D receptor mutation     

See Refs. 1, 7, 14, 18–21, 23, 26, 27, 30–36, 39, 40, 43–46, 53, 70, 78–81.

Diseases of Hypophosphatemia

Three primary mechanisms of hypophosphatemia exist: increased renal excretion, decreased intestinal absorption, and movement of phosphate from the extracellular to intracellular compartments. The initial step in diagnosis involves determining whether hypophosphatemia is renal or nonrenal. Renal hypophosphatemia can be FGF23-mediated or non-FGF23-mediated. Some renal or gastrointestinal mechanisms for hypophosphatemia may cause either acute or chronic hypophosphatemia. Movement of phosphate into cells can cause acute hypophosphatemia, usually following a period of phosphate depletion. Multiple mechanisms may coincide in some patients.

Renal

The classic, and most common, inherited renal phosphate wasting disorder is XLH. XLH usually presents with typical signs of rickets in young children, accompanied by hypophosphatemia, inappropriately low or normal 1,25OHD, and low TmP/GFR. The inheritance pattern is X-linked dominant. Consequently, both males and females are affected, and there is no male-to-male transmission of the disease. However, severity of disease varies widely, even within a kindred (10). Poor linear growth is common, often despite adequate medical treatment. Some data suggest that earlier diagnosis and treatment improve height outcomes (11). However, patients often still require orthopedic surgical procedures (such as femoral or tibial osteotomies) to modulate the progression of lower extremity deformities. Additional features of XLH include frequent dental abscesses and the development of enthesopathy (calcification of tendons and ligaments). Enthesopathy frequently causes joint stiffness and can be the most limiting feature of the disease in adulthood. Adults commonly have significant osteoarthritis with resulting joint pain (12).

XLH is caused by inactivating mutations in PHEX (13), which lead to increased bone expression of the phosphate-regulating hormone FGF23 (14). FGF23 enters the circulation and acts primarily at renal tubular FGF receptors, in conjunction with the coreceptor klotho (15), decreasing proximal renal tubular phosphate reabsorption and 1,25OHD production, and also increasing vitamin D degradation (16). Although these effects on phosphate transport and 1,25OHD metabolism occur in the proximal tubule, FGF23 signaling appears to start in the distal tubule (17). Mechanisms translating the initial distal tubule signal to proximal tubule effect have not been determined. Nevertheless, FGF23 excess causes the biochemical phenotype of XLH. Less common autosomal recessive forms of hypophosphatemic rickets are caused by inactivating mutations in both dentin matrix protein 1 (DMP1) and in ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), both with hypophosphatemia due to excess FGF23 (18–20).

Mutations in FGF23 resulting in impaired cleavage cause FGF23 excess in autosomal dominant hypophosphatemic rickets (ADHR) (21–23). These patients have an interesting clinical phenotype characterized by variability in age of clinically evident disease and incomplete penetrance (24). Hypophosphatemic rickets may develop during childhood, like XLH patients. However, a significant number of subjects from the same kindreds had a delayed onset of phenotype, with normophosphatemia and normal growth without rickets during childhood, to be followed by onset of hypophosphatemic osteomalacia in adolescence or adulthood. Most ADHR patients with delayed onset have been women (23–25). Furthermore, some subjects have later normalized their serum phosphate and stopped therapy. These changes over time are due to alterations in FGF23 (23) concentrations, suggesting that ADHR patients are intermittently regulating FGF23 and phosphate normally.

FGF23-mediated hypophosphatemia also occurs associated with focal sites of abnormal FGF23 production, including fibrous dysplasia (FD) of bone, linear sebaceous nevus syndrome, and tumor-induced osteomalacia (TIO). FD can be associated with café au lait macules and precocious puberty or other endocrine hyperfunction in McCune-Albright syndrome. Consequently, patients with FD should be tested for additional endocrine abnormalities. Phosphate wasting and FGF23 concentration correlates with the total amount of FD lesions (26). Linear sebaceous nevus syndrome is a neuroectodermal disorder associated with seizures, developmental defects, and cutaneous lesions producing FGF23 (27).

Acquired chronic renal phosphate wasting should prompt consideration of TIO, although ADHR, FD, and some types of Fanconi syndrome may also present after childhood. The biochemical phenotype of TIO is identical to that of XLH, but may be more severe. Many tumor types have been reported, including some malignant tumors, but in general these tumors are small, benign, and can be located anywhere in the body. The majority of tumors are classified as phosphaturic mesenchymal tumors of a mixed connective tissue type (28). Multiple phosphatonins have been implicated in TIO, but FGF23 is the most consistent and well-characterized (29–32). If measurement can be made, elevated circulating FGF23 concentration is a sensitive indicator of TIO (30). However, it is not specific because many other disorders involve high FGF23 concentrations.

Hypophosphatemia occurring after renal transplant is caused by prolonged excess FGF23 production acquired during chronic kidney disease (33). Excess FGF23 production gradually resolves over time but may require treatment with phosphate and calcitriol with gradual tapering of doses until the elevated FGF23 resolves.

Some renal disorders associated with phosphate wasting are not FGF23-mediated, but are due to direct tubular abnormalities. Hereditary hypophosphatemic rickets with hypercalciuria (HHRH) is due to mutations in NaPi2c, a sodium-dependent phosphate cotransporter in the renal proximal tubule (34). Contrary to XLH patients, those with HHRH develop an appropriate increase in 1,25OHD and resultant hyperabsorption of calcium, hypercalciuria, and a propensity to nephrolithiasis. Fanconi syndrome is due to a variety of drugs, toxins, or genetic causes, resulting in variable renal losses of phosphate, calcium, and other ions; glucosuria; and aminoaciduria. Some genetic causes of hypophosphatemia due to Fanconi syndrome include mutations in chloride channel 5 (Dent's disease), sodium phosphate cotransporter 2a (NaPi2a), and cystinosin (CTNS, causing nephropathic cystinosis) (35–37).

Posthepatic resection hypophosphatemia is common and can last several days, but the cause is unclear. Although renal phosphate losses occur, this transient hypophosphatemia does not appear to be mediated by either FGF23 or PTH (38).

Nonrenal

Impaired phosphate intake or gastrointestinal absorption causes acute or chronic hypophosphatemia, but shifts from the extracellular compartment to the intracellular compartment cause acute hypophosphatemia. Frequently acute hypophosphatemia is nonrenal in origin, but it often involves a combination of factors.

Dietary phosphate deficiency is not usually an isolated deficiency, but is usually combined with vitamin D and other nutritional deficiencies. Premature infants require higher mineral intake than older infants for skeletal mineralization and thus require additional mineral supplementation, especially if fed with human milk (39). Acutely or chronically ill patients often have impaired nutritional intake or gastrointestinal absorption predisposing to hypophosphatemia. Antacids or other phosphate-binding agents impair absorption, causing acute or chronic hypophosphatemia (1). Parenteral nutrition may contribute to hypophosphatemia if insufficient phosphate is provided.

Any conditions resulting in poor overall nutrition, such as alcoholism, anorexia, severe malabsorption, and starvation, can cause phosphate depletion. Sometimes serum phosphate is normal, despite overall depletion before nutritional intervention. During refeeding, phosphate is shifted into cells for glucose utilization, resulting in hypophosphatemia. Patients with refeeding syndrome are also at risk for hypokalemia and hypomagnesemia. Likewise, phosphate depletion results from hyperglycemia-induced osmotic diuresis in the diabetic ketoacidosis patient. Once insulin administration begins, glucose utilization and cellular phosphate uptake increase and hypophosphatemia develops (1).

Similarly, during salicylate poisoning or rapid mechanical ventilation, phosphate moves into cells due to respiratory alkalosis (5, 40). Anxiety-induced hyperventilation may likewise cause transient hypophosphatemia (41). These acute forms of hypophosphatemia quickly improve with resolution of respiratory alkalosis.

Multiple drugs and toxins induce acute or chronic hypophosphatemia through a variety of mechanisms: isolated phosphaturia, Fanconi syndrome, intestinal phosphate binding, or intracellular uptake of phosphate (Table 1). In one study, 82% of inpatient acute hypophosphatemia were attributed to medications (42). Some forms of iv iron cause hypophosphatemia and osteomalacia due to increased FGF23 concentrations (43–45). Increased urinary excretion of phosphate may complicate diuretic and corticosteroid use. Mannitol can cause a pseudohypophosphatemia due to an assay artifact, but it may also have a mild phosphaturic effect (1). Although phosphate concentrations increase during advanced chronic kidney disease, daily or continuous hemodialysis and phosphate binders can sometimes induce hypophosphatemia (46).

Evaluation

Physical examination

A comprehensive physical examination should identify consequences of hypophosphatemia and clues to an underlying cause. Special focus should be on the musculoskeletal exam, looking for signs of weakness, pathological fractures or pseudofractures, and skeletal deformities. Bone pain may be present, but severe muscle pain may indicate rhabdomyolysis. In children, rachitic features should be noted, and in adults rachitic features suggest chronic hypophosphatemia since childhood. Short stature with increased upper to lower segment ratio also suggests previous childhood rickets, even without leg deformities. Decreased range of motion at the spine, hips, and other large joints can indicate calcified entheses, a common feature in adults with XLH. Facial asymmetry or deformation of long bones may be signs of underlying FD. Maxillary bones are common sites of FD, whereas the sinuses are a common location for tumors causing TIO. If TIO is suspected, a thorough examination for palpable soft tissue masses should be performed. Hepatomegaly may suggest either an underlying tumor or chronic alcoholism. Skin findings such as café au lait macules (FD and McCune Albright syndrome) and linear sebaceous nevi should be noted.

Laboratory assessment

Laboratory assessment should establish the underlying cause of hypophosphatemia (Table 2). During treatment of diabetic ketoacidosis, or refeeding, or after known medications, the cause of hypophosphatemia is usually clear. Otherwise, the first step is to determine whether hypophosphatemia is renal or nonrenal in origin. This requires assessment of the TmP/GFR (Fig. 1) (47). Simultaneous collection of fasting second morning void urine and blood is usually sufficient; if the results are unclear, however, a specific fasting 2-h morning urine collection should be performed, with blood sampling halfway through the collection. The tubular reabsorption of phosphate is calculated and is used with serum phosphate to determine the TmP/GFR from a nomogram (Fig. 1). The TmP/GFR normal range approximates the same numerical range as the age-appropriate serum phosphate concentration in milligrams per deciliter. If both serum phosphate and TmP/GFR are low, this indicates inappropriate renal phosphate wasting. A normal (or high) TmP/GFR indicates renal conservation of phosphate, and hence a nonrenal cause of hypophosphatemia.

Table 2.

Expected laboratory values in the untreated state for selected causes of hypophosphatemia

Serum phosphateSerum calciumSerum ALPSerum PTHSerum 25OHDSerum 1,25OHDSerum FGF23TmP/GFRUrine calcium
Renal hypophosphatemia (TmP/GFR low) 
FGF23-mediated 
XLH, ADHR, ARHR, TIO, FD, postrenal transplant  ↓  ⇆  ↑  ⇆, ↑  ⇆  ⇆, ↓  ↑  ↓  ⇆, ↓ 
Non-FGF23-mediated 
Hyperparathyroidism  ↓  ↑  ⇆, ↑  ↑  ⇆  ↑  ⇆, ↑  ↓  ↓, ⇆, ↑
HHRH (NPT2c)  ↓  ↑  ↓, ⇆  ⇆  ↑  ↓  ↓  ↑ 
Posthepatic resection hypophosphatemia  ↓  ⇆  ⇆, ↑  ⇆  ⇆  ⇆  ↓  ↓  ⇆ 
Diuretics (acetazolamide, thiazides, loop diuretics)  ↓  ⇆, ↑  ⇆  ⇆  ⇆  ⇆  ↓  ↓  ⇆ 
Fanconi syndrome↓  ⇆, ↓  ⇆, ↑  ⇆, ↑  ⇆  ⇆, ↓  ↓  ↓  ↑ 
Nonrenal hypophosphatemia(TmP/GFR normal or high) 
Impaired intestinal absorption or intake 
Impaired dietary intake or malabsorption↓  ⇆, ↓  ⇆, ↑  ⇆, ↑  ⇆, ↓  ↓, ⇆, ↑  ↓  ↑  ↓ 
Phosphate bindersd,↓  ⇆, ↑  ⇄, ↑  ⇆  ⇆  ↑  ↓  ↑  ↓ 
Intracellular uptake 
Refeeding syndrome  ↓  ⇆  ⇆  ⇆  ⇆, ↓  ↓, ⇆, ↑  ↓  ↑  ↓ 

Serum phosphateSerum calciumSerum ALPSerum PTHSerum 25OHDSerum 1,25OHDSerum FGF23TmP/GFRUrine calcium
Renal hypophosphatemia (TmP/GFR low) 
FGF23-mediated 
XLH, ADHR, ARHR, TIO, FD, postrenal transplant  ↓  ⇆  ↑  ⇆, ↑  ⇆  ⇆, ↓  ↑  ↓  ⇆, ↓ 
Non-FGF23-mediated 
Hyperparathyroidism  ↓  ↑  ⇆, ↑  ↑  ⇆  ↑  ⇆, ↑  ↓  ↓, ⇆, ↑
HHRH (NPT2c)  ↓  ↑  ↓, ⇆  ⇆  ↑  ↓  ↓  ↑ 
Posthepatic resection hypophosphatemia  ↓  ⇆  ⇆, ↑  ⇆  ⇆  ⇆  ↓  ↓  ⇆ 
Diuretics (acetazolamide, thiazides, loop diuretics)  ↓  ⇆, ↑  ⇆  ⇆  ⇆  ⇆  ↓  ↓  ⇆ 
Fanconi syndrome↓  ⇆, ↓  ⇆, ↑  ⇆, ↑  ⇆  ⇆, ↓  ↓  ↓  ↑ 
Nonrenal hypophosphatemia(TmP/GFR normal or high) 
Impaired intestinal absorption or intake 
Impaired dietary intake or malabsorption↓  ⇆, ↓  ⇆, ↑  ⇆, ↑  ⇆, ↓  ↓, ⇆, ↑  ↓  ↑  ↓ 
Phosphate bindersd,↓  ⇆, ↑  ⇄, ↑  ⇆  ⇆  ↑  ↓  ↑  ↓ 
Intracellular uptake 
Refeeding syndrome  ↓  ⇆  ⇆  ⇆  ⇆, ↓  ↓, ⇆, ↑  ↓  ↑  ↓ 

ALP, Alkaline phosphatase; ↓, below normal range; ⇆, within normal range; ↑, above normal range.

a

Urine calcium depends on severity of hyperparathyroidism.

b

In HHRH, serum calcium may be high normal, and PTH may be low.

c

Urinary wasting of other electrolytes, glucose, and amino acids.

d

Serum and urine calcium, and serum PTH may be variable. In isolated dietary phosphate deficiency or isolated phosphate malabsorption (most commonly with phosphate binders) 1,25OHD may be up-regulated, leading to increased calcium absorption and potentially hypercalciuria and hypercalcemia.

e

In chronic kidney disease (often treated with phosphate binders), calcium and alkaline phosphatase may be low, and phosphorus, FGF23, and PTH are usually high.

Table 2.

Expected laboratory values in the untreated state for selected causes of hypophosphatemia

Serum phosphateSerum calciumSerum ALPSerum PTHSerum 25OHDSerum 1,25OHDSerum FGF23TmP/GFRUrine calcium
Renal hypophosphatemia (TmP/GFR low) 
FGF23-mediated 
XLH, ADHR, ARHR, TIO, FD, postrenal transplant  ↓  ⇆  ↑  ⇆, ↑  ⇆  ⇆, ↓  ↑  ↓  ⇆, ↓ 
Non-FGF23-mediated 
Hyperparathyroidism  ↓  ↑  ⇆, ↑  ↑  ⇆  ↑  ⇆, ↑  ↓  ↓, ⇆, ↑
HHRH (NPT2c)  ↓  ↑  ↓, ⇆  ⇆  ↑  ↓  ↓  ↑ 
Posthepatic resection hypophosphatemia  ↓  ⇆  ⇆, ↑  ⇆  ⇆  ⇆  ↓  ↓  ⇆ 
Diuretics (acetazolamide, thiazides, loop diuretics)  ↓  ⇆, ↑  ⇆  ⇆  ⇆  ⇆  ↓  ↓  ⇆ 
Fanconi syndrome↓  ⇆, ↓  ⇆, ↑  ⇆, ↑  ⇆  ⇆, ↓  ↓  ↓  ↑ 
Nonrenal hypophosphatemia(TmP/GFR normal or high) 
Impaired intestinal absorption or intake 
Impaired dietary intake or malabsorption↓  ⇆, ↓  ⇆, ↑  ⇆, ↑  ⇆, ↓  ↓, ⇆, ↑  ↓  ↑  ↓ 
Phosphate bindersd,↓  ⇆, ↑  ⇄, ↑  ⇆  ⇆  ↑  ↓  ↑  ↓ 
Intracellular uptake 
Refeeding syndrome  ↓  ⇆  ⇆  ⇆  ⇆, ↓  ↓, ⇆, ↑  ↓  ↑  ↓ 

Serum phosphateSerum calciumSerum ALPSerum PTHSerum 25OHDSerum 1,25OHDSerum FGF23TmP/GFRUrine calcium
Renal hypophosphatemia (TmP/GFR low) 
FGF23-mediated 
XLH, ADHR, ARHR, TIO, FD, postrenal transplant  ↓  ⇆  ↑  ⇆, ↑  ⇆  ⇆, ↓  ↑  ↓  ⇆, ↓ 
Non-FGF23-mediated 
Hyperparathyroidism  ↓  ↑  ⇆, ↑  ↑  ⇆  ↑  ⇆, ↑  ↓  ↓, ⇆, ↑
HHRH (NPT2c)  ↓  ↑  ↓, ⇆  ⇆  ↑  ↓  ↓  ↑ 
Posthepatic resection hypophosphatemia  ↓  ⇆  ⇆, ↑  ⇆  ⇆  ⇆  ↓  ↓  ⇆ 
Diuretics (acetazolamide, thiazides, loop diuretics)  ↓  ⇆, ↑  ⇆  ⇆  ⇆  ⇆  ↓  ↓  ⇆ 
Fanconi syndrome↓  ⇆, ↓  ⇆, ↑  ⇆, ↑  ⇆  ⇆, ↓  ↓  ↓  ↑ 
Nonrenal hypophosphatemia(TmP/GFR normal or high) 
Impaired intestinal absorption or intake 
Impaired dietary intake or malabsorption↓  ⇆, ↓  ⇆, ↑  ⇆, ↑  ⇆, ↓  ↓, ⇆, ↑  ↓  ↑  ↓ 
Phosphate bindersd,↓  ⇆, ↑  ⇄, ↑  ⇆  ⇆  ↑  ↓  ↑  ↓ 
Intracellular uptake 
Refeeding syndrome  ↓  ⇆  ⇆  ⇆  ⇆, ↓  ↓, ⇆, ↑  ↓  ↑  ↓ 

ALP, Alkaline phosphatase; ↓, below normal range; ⇆, within normal range; ↑, above normal range.

a

Urine calcium depends on severity of hyperparathyroidism.

b

In HHRH, serum calcium may be high normal, and PTH may be low.

c

Urinary wasting of other electrolytes, glucose, and amino acids.

d

Serum and urine calcium, and serum PTH may be variable. In isolated dietary phosphate deficiency or isolated phosphate malabsorption (most commonly with phosphate binders) 1,25OHD may be up-regulated, leading to increased calcium absorption and potentially hypercalciuria and hypercalcemia.

e

In chronic kidney disease (often treated with phosphate binders), calcium and alkaline phosphatase may be low, and phosphorus, FGF23, and PTH are usually high.

Fig. 1.

Nomogram for determining TmP/GFR. The tubular reabsorption of phosphate (TRP) is calculated using the formula: 1 − [(urine phosphate * serum creatinine)/(serum phosphate * urine creatinine)]. Left axis, PO4 indicates phosphate in mg/dl to the outside of the axis and mmol/l to the inside of the axis. Right axis, TmPO4/GFR is the renal threshold phosphate concentration in mg/dl to the outside of the axis and in mmol/l to the inside of the axis. CPO4 is the clearance of phosphate. Ccreat is the clearance of creatinine. [Reprinted from R. J. Walton and O. L. Bijvoet: Nomogram for derivation of renal threshold phosphate concentration. Lancet 306:309–310, 1975 (47), with permission. © Elsevier.]

In chronic hypophosphatemia, osteomalacia develops and the total (or bone-specific) alkaline phosphatase is usually in the high or high-normal range. Concomitant hypercalcemia during hypophosphatemia suggests hyperparathyroidism, whereas hypocalcemia suggests vitamin D deficiency or other abnormality in vitamin D metabolism. 25OHD should be measured because vitamin D deficiency may accompany poor nutrition and phosphate deficiency. In FGF23-mediated disorders, 1,25OHD will be inappropriately low or normal during hypophosphatemia, but in other disorders, the physiological response to hypophosphatemia results in elevated 1,25OHD concentrations. Measuring PTH allows for determination of hyperparathyroidism, although patients with FGF23-mediated disorders may also develop hyperparathyroidism (48, 49). FGF23 measurement is not yet routinely available in clinical practice but is potentially useful in evaluating chronic hypophosphatemia (29).

Assessing urine calcium and creatinine in either a fasting or 24-h specimen allows for detection of hypercalciuria that can be seen in Fanconi syndrome, hypercalcemic hyperparathyroidism, and some inherited forms of hypophosphatemia. Aminoaciduria, proteinuria, microglobulinuria, and glucosuria are also indicators of Fanconi syndrome.

Imaging

For most acute hypophosphatemia, imaging is not necessary unless required to evaluate neurological, cardiac, or respiratory complications. For chronic hypophosphatemia, imaging is performed to evaluate both causes and consequences of hypophosphatemia.

Plain radiographs of areas of deformity noted on physical examination or of specific sites of pain should be performed to detect signs of rickets, FD, or pseudofractures. With severe osteomalacia, 99technetium bone scintigraphy may demonstrate increased uptake at multiple areas, including ribs and pseudofractures. FD lesions may also be detected by bone scintigraphy (26). Computed tomography or magnetic resonance imaging is used to more fully characterize craniofacial or spinal FD lesions and evaluate possible neurovascular impingement.

When TIO is suspected, imaging should be directed by thorough physical examination because tumors may be palpable. Tumors causing TIO are often difficult to find. A wide variety of imaging techniques have been used to detect causative tumors, including plain radiographs, computed tomography, magnetic resonance imaging, octreotide scanning, and combined positron emission tomography/computed tomography (50–56). Whole body imaging techniques may offer an advantage. However, there appears to be a reporting bias for positive detection of tumors with various modalities. In our experience, sensitivity of each method is limited. A tumor is probably located only about half the time, despite multiple imaging techniques (30, 31). Furthermore, sometimes an identified lesion represents false positivity and is not causative of hypophosphatemia. Sometimes after multiple negative imaging studies, tumors are subsequently identified when tests are repeated after a few years.

Selective venous sampling for FGF23 concentrations has identified 18 tumors in five different reports (57–61). An estimated sensitivity of 87% and specificity of 71% were reported for a minimum intact FGF23 ratio of 1.6 between the maximum FGF23 concentration and the mean of the other sites sampled (61). However, false-positive results were noted, and venous sampling was nondiagnostic for six patients without otherwise identifiable lesions on comprehensive imaging (1, 61). Although this technique has potential, we recommend that it be considered experimental.

Hypophosphatemic osteomalacia is sometimes discovered as low bone density on dual-energy x-ray absorptiometry scanning. This is more common with acquired hypophosphatemia, and treatment rapidly improves bone density. However, although the mechanism is uncertain, patients with XLH often have mildly increased bone density, despite active osteomalacia (62).

Bone biopsy for osteomalacia is not typically needed but shows increased unmineralized osteoid and delayed mineralization rate. Biopsy of patients with XLH shows periosteocytic halos that are not present in TIO patients (55).

Management

Treatment will alter some diagnostic tests, so it is useful to obtain the proper biochemical testing to identify renal or nonrenal hypophosphatemia before initiating treatment. Treatment should address the underlying cause when possible, removing causative drugs or addressing dietary deficiencies. Acute severe hypophosphatemia in hospitalized patients may contribute to respiratory or hemodynamic instability, and a 2- to 4-fold increase in mortality is reported in prospective and retrospective studies (63, 64), while repletion can improve hemodynamic or respiratory instability.

When treating acute hypophosphatemia, the underlying cause must be addressed when possible. Intravenous phosphate is appropriate in the acute setting: when critically ill, when enteral intake is impaired, or when oral phosphate is not tolerated, especially when serum phosphate is less than 1.5 mg/dl. When hypophosphatemia is an expected complication of medical treatment, such as in refeeding syndrome, chronic alcoholic patients, or diabetic ketoacidosis, phosphate is added to maintenance iv fluids to prevent or treat hypophosphatemia, usually in the form of potassium phosphate.

Intravenous phosphate should be used cautiously. Treatment of severe acute hypophosphatemia is based on small uncontrolled adult studies with only 10 to 16 patients with serum phosphate below 1.5 mg/dl per study (65–68). Varying regimens are published (Table 3), which are not validated in children. However, the response to iv phosphate is highly variable and not easily predicted by initial levels. When providing iv phosphate, serum calcium, phosphate, potassium, magnesium, and creatinine should be closely monitored (at least every 6 h), and telemetry is recommended. The most significant risks of iv phosphate are acute severe life-threatening hypocalcemia, with tetany, seizures, electrocardiogram changes and shock, and overtreatment resulting in hyperphosphatemia and hyperkalemia (because of potassium phosphate formulations). Ectopic mineralization may occur with aggressive repletion. Intravenous phosphate should not be given to hypocalcemic patients. Concomitant low calcium and phosphate suggests vitamin D deficiency, and such patients should be managed with repletion of vitamin D or calcitriol. Patients with renal failure are also at higher risk of complications from iv phosphate. In general, oral phosphate is safer and is the preferred route in the stable patient with acute or chronic hypophosphatemia. However, hypocalcemia may still occur during aggressive oral phosphate repletion.

Table 3.

Treatment of hypophosphatemia

First author (Ref.)Dose of elemental phosphorusMonitoring
Acute hypophosphatemia iv phosphate regimens (serum phosphate <1.5 mg/dl)a,b,Rosen et al. (68)  15 mmol (464 mg) in 100 ml 0.9% saline over 2-h bolus, repeated after 6 h if needed [maximum 45 mmol (1393 mg) in 24 h]  Calcium, phosphate, potassium, magnesium, creatinine every 6 h; electrocardiogram monitoring/telemetry 
Vannatta et al. (67)  0.32 mmol/kg (9.9 mg/kg) infused over 12 h and repeated every 12 h until serum phosphate was >2 mg/dl 
Clark et al. (66)  0.64 mmol/kg (19.8 mg/kg) over 8–12 h 
  Taylor et al. (adapted) (65)  If phosphate 1.0–1.7 mg/dl, give 0.4 mmol/kg (12.4 mg/kg) over 6 h, maximum dose 40 mmol (1238 mg). If phosphate <1 mg/dl, give 0.5 mmol/kg (15.5 mg/kg) over 6 h, maximum dose 50 mmol (1548 mg)   
Oral phosphate saltsb,Shiber et al. (4), Miller et al. (82)  30–40 mg/kg per day in four or five divided doses  Calcium, phosphate, potassium, magnesium, creatinine every 12–24 h 
Vitamin D    800 to 1000 units daily (may require more if deficient)   
Refeeding syndromeb,Stanga et al. (83)  0.5–0.8 mmol/kg per day (15–25 mg/kg per day) in iv fluids  Calcium, phosphate, potassium, magnesium, creatinine every 12–24 h 
Chronic hypophosphatemia (especially renal phosphate wasting)     
Phosphate saltsCarpenter et al. (70)  20–40 mg/kg per day divided into four doses  Calcium, phosphate, potassium, creatinine monthly until stable, then every 3 months with alkaline phosphatase, PTH every 6 to 12 months, renal ultrasound every 1–2 yr 
CalcitriolCarpenter et al. (70)  20–30 ng/kg per day divided into two doses   

First author (Ref.)Dose of elemental phosphorusMonitoring
Acute hypophosphatemia iv phosphate regimens (serum phosphate <1.5 mg/dl)a,b,Rosen et al. (68)  15 mmol (464 mg) in 100 ml 0.9% saline over 2-h bolus, repeated after 6 h if needed [maximum 45 mmol (1393 mg) in 24 h]  Calcium, phosphate, potassium, magnesium, creatinine every 6 h; electrocardiogram monitoring/telemetry 
Vannatta et al. (67)  0.32 mmol/kg (9.9 mg/kg) infused over 12 h and repeated every 12 h until serum phosphate was >2 mg/dl 
Clark et al. (66)  0.64 mmol/kg (19.8 mg/kg) over 8–12 h 
  Taylor et al. (adapted) (65)  If phosphate 1.0–1.7 mg/dl, give 0.4 mmol/kg (12.4 mg/kg) over 6 h, maximum dose 40 mmol (1238 mg). If phosphate <1 mg/dl, give 0.5 mmol/kg (15.5 mg/kg) over 6 h, maximum dose 50 mmol (1548 mg)   
Oral phosphate saltsb,Shiber et al. (4), Miller et al. (82)  30–40 mg/kg per day in four or five divided doses  Calcium, phosphate, potassium, magnesium, creatinine every 12–24 h 
Vitamin D    800 to 1000 units daily (may require more if deficient)   
Refeeding syndromeb,Stanga et al. (83)  0.5–0.8 mmol/kg per day (15–25 mg/kg per day) in iv fluids  Calcium, phosphate, potassium, magnesium, creatinine every 12–24 h 
Chronic hypophosphatemia (especially renal phosphate wasting)     
Phosphate saltsCarpenter et al. (70)  20–40 mg/kg per day divided into four doses  Calcium, phosphate, potassium, creatinine monthly until stable, then every 3 months with alkaline phosphatase, PTH every 6 to 12 months, renal ultrasound every 1–2 yr 
CalcitriolCarpenter et al. (70)  20–30 ng/kg per day divided into two doses   

a

Intravenous regimens are listed from four adult studies; caution should be used if applying to children. Intravenous phosphate should be avoided in hypocalcemic or hypercalcemic patients. Intravenous doses of phosphate over 30 mmol should be given in a central venous catheter.

b

Severe hypocalcemia may result from aggressive iv or oral phosphate repletion. Impaired kidney function may increase the risk of hypocalcemia.

c

Potassium phosphate salts should not be used if the patient is hyperkalemic or has impaired kidney function.

d

Primary renal phosphate wasting disorders, especially FGF23-mediated disorders such as XLH, usually require treatment with both calcitriol and phosphate. Phosphate is not used alone (without calcitriol) in XLH or when PTH is elevated because this can cause or worsen hyperparathyroidism.

e

If hypercalciuria is present at diagnosis, this should be confirmed, and if persistent, calcitriol should be avoided because calcitriol treatment will increase calciuria. Hypercalciuria in a previously untreated patient may indicate a different type of phosphate wasting disorder, such as HHRH, which is not treated with calcitriol.

Table 3.

Treatment of hypophosphatemia

First author (Ref.)Dose of elemental phosphorusMonitoring
Acute hypophosphatemia iv phosphate regimens (serum phosphate <1.5 mg/dl)a,b,Rosen et al. (68)  15 mmol (464 mg) in 100 ml 0.9% saline over 2-h bolus, repeated after 6 h if needed [maximum 45 mmol (1393 mg) in 24 h]  Calcium, phosphate, potassium, magnesium, creatinine every 6 h; electrocardiogram monitoring/telemetry 
Vannatta et al. (67)  0.32 mmol/kg (9.9 mg/kg) infused over 12 h and repeated every 12 h until serum phosphate was >2 mg/dl 
Clark et al. (66)  0.64 mmol/kg (19.8 mg/kg) over 8–12 h 
  Taylor et al. (adapted) (65)  If phosphate 1.0–1.7 mg/dl, give 0.4 mmol/kg (12.4 mg/kg) over 6 h, maximum dose 40 mmol (1238 mg). If phosphate <1 mg/dl, give 0.5 mmol/kg (15.5 mg/kg) over 6 h, maximum dose 50 mmol (1548 mg)   
Oral phosphate saltsb,Shiber et al. (4), Miller et al. (82)  30–40 mg/kg per day in four or five divided doses  Calcium, phosphate, potassium, magnesium, creatinine every 12–24 h 
Vitamin D    800 to 1000 units daily (may require more if deficient)   
Refeeding syndromeb,Stanga et al. (83)  0.5–0.8 mmol/kg per day (15–25 mg/kg per day) in iv fluids  Calcium, phosphate, potassium, magnesium, creatinine every 12–24 h 
Chronic hypophosphatemia (especially renal phosphate wasting)     
Phosphate saltsCarpenter et al. (70)  20–40 mg/kg per day divided into four doses  Calcium, phosphate, potassium, creatinine monthly until stable, then every 3 months with alkaline phosphatase, PTH every 6 to 12 months, renal ultrasound every 1–2 yr 
CalcitriolCarpenter et al. (70)  20–30 ng/kg per day divided into two doses   

First author (Ref.)Dose of elemental phosphorusMonitoring
Acute hypophosphatemia iv phosphate regimens (serum phosphate <1.5 mg/dl)a,b,Rosen et al. (68)  15 mmol (464 mg) in 100 ml 0.9% saline over 2-h bolus, repeated after 6 h if needed [maximum 45 mmol (1393 mg) in 24 h]  Calcium, phosphate, potassium, magnesium, creatinine every 6 h; electrocardiogram monitoring/telemetry 
Vannatta et al. (67)  0.32 mmol/kg (9.9 mg/kg) infused over 12 h and repeated every 12 h until serum phosphate was >2 mg/dl 
Clark et al. (66)  0.64 mmol/kg (19.8 mg/kg) over 8–12 h 
  Taylor et al. (adapted) (65)  If phosphate 1.0–1.7 mg/dl, give 0.4 mmol/kg (12.4 mg/kg) over 6 h, maximum dose 40 mmol (1238 mg). If phosphate <1 mg/dl, give 0.5 mmol/kg (15.5 mg/kg) over 6 h, maximum dose 50 mmol (1548 mg)   
Oral phosphate saltsb,Shiber et al. (4), Miller et al. (82)  30–40 mg/kg per day in four or five divided doses  Calcium, phosphate, potassium, magnesium, creatinine every 12–24 h 
Vitamin D    800 to 1000 units daily (may require more if deficient)   
Refeeding syndromeb,Stanga et al. (83)  0.5–0.8 mmol/kg per day (15–25 mg/kg per day) in iv fluids  Calcium, phosphate, potassium, magnesium, creatinine every 12–24 h 
Chronic hypophosphatemia (especially renal phosphate wasting)     
Phosphate saltsCarpenter et al. (70)  20–40 mg/kg per day divided into four doses  Calcium, phosphate, potassium, creatinine monthly until stable, then every 3 months with alkaline phosphatase, PTH every 6 to 12 months, renal ultrasound every 1–2 yr 
CalcitriolCarpenter et al. (70)  20–30 ng/kg per day divided into two doses   

a

Intravenous regimens are listed from four adult studies; caution should be used if applying to children. Intravenous phosphate should be avoided in hypocalcemic or hypercalcemic patients. Intravenous doses of phosphate over 30 mmol should be given in a central venous catheter.

b

Severe hypocalcemia may result from aggressive iv or oral phosphate repletion. Impaired kidney function may increase the risk of hypocalcemia.

c

Potassium phosphate salts should not be used if the patient is hyperkalemic or has impaired kidney function.

d

Primary renal phosphate wasting disorders, especially FGF23-mediated disorders such as XLH, usually require treatment with both calcitriol and phosphate. Phosphate is not used alone (without calcitriol) in XLH or when PTH is elevated because this can cause or worsen hyperparathyroidism.

e

If hypercalciuria is present at diagnosis, this should be confirmed, and if persistent, calcitriol should be avoided because calcitriol treatment will increase calciuria. Hypercalciuria in a previously untreated patient may indicate a different type of phosphate wasting disorder, such as HHRH, which is not treated with calcitriol.

Management of chronic hypophosphatemia depends on the underlying cause. Avoidance of phosphate binders or other causative medications and specific treatment of the underlying cause is appropriate. In Fanconi syndrome, careful replacement with phosphate and calcium may be required. HHRH is typically treated with phosphate, but not calcitriol, because 1,25OHD is appropriately elevated in these patients, resulting in hypercalciuria.

The standard medical treatment of FGF23-mediated disorders (XLH, FD, TIO, ADHR, etc.) is based on current knowledge for treating XLH with calcitriol and phosphate. More thorough discussions of which patients should be treated are available elsewhere (69, 70). Almost all children with XLH require therapy, although many still require surgery to correct lower extremity deformities. Decisions about treating adults with XLH are much more complex, but patients with stress fractures and bone pain should be considered for therapy after full discussion of risks. Joint problems in XLH are not improved by current therapy (69).

Current medical therapy for FGF23-mediated disorders consists of attempting to replete the consequences of FGF23 excess. No current treatment alters the effect of FGF23 on the kidney. Because FGF23 excess inhibits both 1,25OHD production and phosphate reabsorption, patients typically receive relatively high doses of phosphate and calcitriol. This standard of care has been demonstrated to improve osteomalacia in XLH (71, 72), but it is based largely on uncontrolled retrospective studies (70). Few studies had any controls (73), and some included historical controls (74, 75).

Potassium- and sodium-phosphate salts are given in doses of elemental phosphorus ranging from 20–40 mg/kg per day in four divided doses (70). Side effects commonly include gastrointestinal distress and diarrhea, which can sometimes be limited by starting at a low dose and titrating up to the target dose. In excessive doses, oral or iv phosphate can cause an acute phosphate nephropathy and renal failure. Calcitriol dosing is usually 20–30 ng/kg per day, although transiently higher doses may be used for several months to speed healing of osteomalacia, followed by decreasing to the above maintenance dose.

This therapy requires close monitoring of serum and urine calcium, phosphate, and creatinine, with careful dose adjustment. Because treatment with phosphate and calcitriol increases the daily urine calcium and phosphate excreted, we do not target consistently normal serum phosphate concentrations. In fact, persistent normal serum phosphate concentrations may be an indication for decreasing the doses. After initial healing of osteomalacia, skeletal calcium uptake decreases and hypercalciuria or hypercalcemia indicates the calcitriol dose should be decreased. Likewise, if serum phosphate becomes elevated, the phosphate and/or the calcitriol dose should be decreased. Although not always elevated (especially in adults with XLH), the alkaline phosphatase will generally decrease during treatment.

Nephrocalcinosis, renal insufficiency, and secondary (sometimes tertiary) hyperparathyroidism can be complications of this therapy (49, 76). Consequently, we recommend monitoring of PTH at least once or twice yearly and renal ultrasounds every 1 or 2 yr in patients being treated with calcitriol and phosphate. Of note, current treatment with calcitriol and phosphate also tends to increase FGF23 concentrations (77).

In TIO, the goal of treatment should be complete resection of the tumor for surgical cure when possible. These tumors also occasionally metastasize, and recurrences may develop years after initial surgical cure. Long-term monitoring of phosphate and alkaline phosphatase is recommended to detect recurrences. If a causative tumor cannot be found and completely resected, TIO patients generally respond dramatically to medical treatment with phosphate and calcitriol as the osteomalacia heals, with resolution of weakness and bone pain, and return to full ambulation.

Back to the Patient

TmP/GFR was low (1.2 mg/dl), indicating renal phosphate wasting. There was no aminoaciduria. PTH was 45 pg/ml. Dual-energy x-ray absorptiometry revealed a T-score of −2.2 at the femoral neck. Intact FGF23 concentration was 97 pg/ml (normal, <70 pg/ml). Treatment with calcitriol and phosphate improved the patient's pain and corrected his alkaline phosphatase. His stress fractures resolved, and he returned to free ambulation. Initial imaging studies failed to reveal a likely source for TIO. Eventually, a positron emission tomography/computed tomography identified a 1.3-cm cortical lesion in the left iliac bone. Resection revealed a spindle cell neoplasm with features consistent with phosphaturic mesenchymal tumor, mixed connective tissue type. He quickly tapered off of calcitriol and phosphate salts; 3 wk after surgery, serum phosphate was 3.9 mg/dl.

Conclusion

Hypophosphatemia is commonly missed due to nonspecific signs and symptoms, but it causes considerable morbidity and can contribute to mortality. Proper diagnosis requires a thorough medication history, family history, examination, and assessment of renal tubular phosphate handling to identify the cause. Imaging studies should be directed at determining complications of hypophosphatemia and identifying potential causes such as TIO or FD. Treatment of hypophosphatemia is determined by the underlying cause. Medical treatment of renal phosphate wasting disorders can dramatically improve weakness and osteomalacia.

Acknowledgments

The authors' work is supported by National Institutes of Health Grants R01 AR042228 (to M.J.E.) and K23 AR057096 (to E.A.I.) from the National Institute of Arthritis and Musculoskeletal and Skin Diseases.

Disclosure Summary: M.J.E. receives royalties from, and is a consultant to, Kyowa Hakko Kirin Pharma, Inc., and E.A.I. participates in a clinical trial with Kyowa Hakko Kirin Pharma, Inc.

Abbreviations

     
  • ADHR

    Autosomal dominant hypophosphatemic rickets

  •  
  • FD

  •  
  • FGF23

    fibroblast growth factor 23

  •  
  • HHRH

    hereditary hypophosphatemic rickets with hypercalciuria

  •  
  • 1,25OHD

  •  
  • 25OHD

  •  
  • TIO

    tumor-induced osteomalacia

  •  
  • TmP/GFR

    tubular maximum reabsorption of phosphate per unit of glomerular filtrate

  •  
  • XLH

    X-linked hypophosphatemia.

References

1.

Liamis

G

,

Milionis

HJ

,

Elisaf

M

2010

Medication-induced hypophosphatemia: a review.

QJM

103

:

449

459

2.

Kruse

K

,

Kracht

U

,

Göpfert

G

1982

Renal threshold phosphate concentration (TmPO4/GFR).

Arch Dis Child

57

:

217

223

3.

Soldin

SJ

,

Hunt

C

,

Hicks

JM

1999

Pediatric reference ranges for phosphorus on the Vitros 500 Analyzer.

Clin Chem

45

:

A22

(abstract)

4.

Shiber

JR

,

Mattu

A

2002

Serum phosphate abnormalities in the emergency department.

J Emerg Med

23

:

395

400

5.

Laaban

JP

,

Marsal

L

,

Waked

M

,

Vuong

TK

,

Rochemaure

J

1990

Seizures related to severe hypophosphataemia induced by mechanical ventilation.

Intensive Care Med

16

:

135

136

6.

Silvis

SE

,

DiBartolomeo

AG

,

Aaker

HM

1980

Hypophosphatemia and neurological changes secondary to oral caloric intake: a variant of hyperalimentation syndrome.

Am J Gastroenterol

73

:

215

222

7.

Silvis

SE

,

Paragas

PD

1972

Paresthesias, weakness, seizures, and hypophosphatemia in patients receiving hyperalimentation.

Gastroenterology

62

:

513

520

8.

Brunelli

SM

,

Goldfarb

S

2007

Hypophosphatemia: clinical consequences and management.

J Am Soc Nephrol

18

:

1999

2003

9.

Econs

MJ

,

Samsa

GP

,

Monger

M

,

Drezner

MK

,

Feussner

JR

1994

X-linked hypophosphatemic rickets: a disease often unknown to affected patients.

Bone Miner

24

:

17

24

10.

Econs

MJ

,

Feussner

JR

,

Samsa

GP

,

Effman

EL

,

Vogler

JB

,

Martinez

S

,

Friedman

NE

,

Quarles

LD

,

Drezner

MK

1991

X-linked hypophosphatemic rickets without “rickets.”

Skeletal Radiol

20

:

109

114

11.

Mäkitie

O

,

Doria

A

,

Kooh

SW

,

Cole

WG

,

Daneman

A

,

Sochett

E

2003

Early treatment improves growth and biochemical and radiographic outcome in X-linked hypophosphatemic rickets.

J Clin Endocrinol Metab

88

:

3591

3597

12.

Reid

IR

,

Hardy

DC

,

Murphy

WA

,

Teitelbaum

SL

,

Bergfeld

MA

,

Whyte

MP

1989

X-linked hypophosphatemia: a clinical, biochemical, and histopathologic assessment of morbidity in adults.

Medicine (Baltimore)

68

:

336

352

13.

Hyp_Consortium

1995

A gene (PEX) with homologies to endopeptidases is mutated in patients with X-linked hypophosphatemic rickets. The HYP Consortium.

Nat Genet

11

:

130

136

14.

Liu

S

,

Guo

R

,

Simpson

LG

,

Xiao

ZS

,

Burnham

CE

,

Quarles

LD

2003

Regulation of fibroblastic growth factor 23 expression but not degradation by PHEX.

J Biol Chem

278

:

37419

37426

15.

Urakawa

I

,

Yamazaki

Y

,

Shimada

T

,

Iijima

K

,

Hasegawa

H

,

Okawa

K

,

Fujita

T

,

Fukumoto

S

,

Yamashita

T

2006

Klotho converts canonical FGF receptor into a specific receptor for FGF23.

Nature

444

:

770

774

16.

Shimada

T

,

Hasegawa

H

,

Yamazaki

Y

,

Muto

T

,

Hino

R

,

Takeuchi

Y

,

Fujita

T

,

Nakahara

K

,

Fukumoto

S

,

Yamashita

T

2004

FGF-23 is a potent regulator of vitamin D metabolism and phosphate homeostasis.

J Bone Miner Res

19

:

429

435

17.

Farrow

EG

,

Davis

SI

,

Summers

LJ

,

White

KE

2009

Initial FGF23-mediated signaling occurs in the distal convoluted tubule.

J Am Soc Nephrol

20

:

955

960

18.

Feng

JQ

,

Ward

LM

,

Liu

S

,

Lu

Y

,

Xie

Y

,

Yuan

B

,

Yu

X

,

Rauch

F

,

Davis

SI

,

Zhang

S

,

Rios

H

,

Drezner

MK

,

Quarles

LD

,

Bonewald

LF

,

White

KE

2006

Loss of DMP1 causes rickets and osteomalacia and identifies a role for osteocytes in mineral metabolism.

Nat Genet

38

:

1310

1315

19.

Levy-Litan

V

,

Hershkovitz

E

,

Avizov

L

,

Leventhal

N

,

Bercovich

D

,

Chalifa-Caspi

V

,

Manor

E

,

Buriakovsky

S

,

Hadad

Y

,

Goding

J

,

Parvari

R

2010

Autosomal-recessive hypophosphatemic rickets is associated with an inactivation mutation in the ENPP1 gene.

Am J Hum Genet

86

:

273

278

20.

Lorenz-Depiereux

B

,

Schnabel

D

,

Tiosano

D

,

Häusler

G

,

Strom

TM

2010

Loss-of-function ENPP1 mutations cause both generalized arterial calcification of infancy and autosomal-recessive hypophosphatemic rickets.

Am J Hum Genet

86

:

267

272

21.

ADHR Consortium

2000

Autosomal dominant hypophosphataemic rickets is associated with mutations in FGF23.

Nat Genet

26

:

345

348

22.

Bai

XY

,

Miao

D

,

Goltzman

D

,

Karaplis

AC

2003

The autosomal dominant hypophosphatemic rickets R176Q mutation in fibroblast growth factor 23 resists proteolytic cleavage and enhances in vivo biological potency.

J Biol Chem

278

:

9843

9849

23.

Imel

EA

,

Hui

SL

,

Econs

MJ

2007

FGF23 concentrations vary with disease status in autosomal dominant hypophosphatemic rickets.

J Bone Miner Res

22

:

520

526

24.

Econs

MJ

,

McEnery

PT

1997

Autosomal dominant hypophosphatemic rickets/osteomalacia: clinical characterization of a novel renal phosphate-wasting disorder.

J Clin Endocrinol Metab

82

:

674

681

25.

Gribaa

M

,

Younes

M

,

Bouyacoub

Y

,

Korbaa

W

,

Ben Charfeddine

I

,

Touzi

M

,

Adala

L

,

Mamay

O

,

Bergaoui

N

,

Saad

A

2010

An autosomal dominant hypophosphatemic rickets phenotype in a Tunisian family caused by a new FGF23 missense mutation.

J Bone Miner Metab

28

:

111

115

26.

Riminucci

M

,

Collins

MT

,

Fedarko

NS

,

Cherman

N

,

Corsi

A

,

White

KE

,

Waguespack

S

,

Gupta

A

,

Hannon

T

,

Econs

MJ

,

Bianco

P

,

Gehron Robey

P

2003

FGF-23 in fibrous dysplasia of bone and its relationship to renal phosphate wasting.

J Clin Invest

112

:

683

692

27.

Hoffman

WH

,

Jueppner

HW

,

Deyoung

BR

,

O'dorisio

MS

,

Given

KS

2005

Elevated fibroblast growth factor-23 in hypophosphatemic linear nevus sebaceous syndrome.

Am J Med Genet A

134

:

233

236

28.

Folpe

AL

,

Fanburg-Smith

JC

,

Billings

SD

,

Bisceglia

M

,

Bertoni

F

,

Cho

JY

,

Econs

MJ

,

Inwards

CY

,

Jan de Beur

SM

,

Mentzel

T

,

Montgomery

E

,

Michal

M

,

Miettinen

M

,

Mills

SE

,

Reith

JD

,

O'Connell

JX

,

Rosenberg

AE

,

Rubin

BP

,

Sweet

DE

,

Vinh

TN

,

Wold

LE

,

Wehrli

BM

,

White

KE

,

Zaino

RJ

,

Weiss

SW

2004

Most osteomalacia-associated mesenchymal tumors are a single histopathologic entity: an analysis of 32 cases and a comprehensive review of the literature.

Am J Surg Pathol

28

:

1

30

29.

Endo

I

,

Fukumoto

S

,

Ozono

K

,

Namba

N

,

Tanaka

H

,

Inoue

D

,

Minagawa

M

,

Sugimoto

T

,

Yamauchi

M

,

Michigami

T

,

Matsumoto

T

2008

Clinical usefulness of measurement of fibroblast growth factor 23 (FGF23) in hypophosphatemic patients. Proposal of diagnostic criteria using FGF23 measurement.

Bone

42

:

1235

1239

30.

Imel

EA

,

Peacock

M

,

Pitukcheewanont

P

,

Heller

HJ

,

Ward

LM

,

Shulman

D

,

Kassem

M

,

Rackoff

P

,

Zimering

M

,

Dalkin

A

,

Drobny

E

,

Colussi

G

,

Shaker

JL

,

Hoogendoorn

EH

,

Hui

SL

,

Econs

MJ

2006

Sensitivity of fibroblast growth factor 23 measurements in tumor-induced osteomalacia.

J Clin Endocrinol Metab

91

:

2055

2061

31.

Jonsson

KB

,

Zahradnik

R

,

Larsson

T

,

White

KE

,

Sugimoto

T

,

Imanishi

Y

,

Yamamoto

T

,

Hampson

G

,

Koshiyama

H

,

Ljunggren

O

,

Oba

K

,

Yang

IM

,

Miyauchi

A

,

Econs

MJ

,

Lavigne

J

,

Jüppner

H

2003

Fibroblast growth factor 23 in oncogenic osteomalacia and X-linked hypophosphatemia.

N Engl J Med

348

:

1656

1663

32.

Yamazaki

Y

,

Okazaki

R

,

Shibata

M

,

Hasegawa

Y

,

Satoh

K

,

Tajima

T

,

Takeuchi

Y

,

Fujita

T

,

Nakahara

K

,

Yamashita

T

,

Fukumoto

S

2002

Increased circulatory level of biologically active full-length FGF-23 in patients with hypophosphatemic rickets/osteomalacia.

J Clin Endocrinol Metab

87

:

4957

4960

33.

Bhan

I

,

Shah

A

,

Holmes

J

,

Isakova

T

,

Gutierrez

O

,

Burnett

SM

,

Jüppner

H

,

Wolf

M

2006

Post-transplant hypophosphatemia: tertiary ‘hyper-phosphatoninism?’

Kidney Int

70

:

1486

1494

34.

Bergwitz

C

,

Roslin

NM

,

Tieder

M

,

Loredo-Osti

JC

,

Bastepe

M

,

Abu-Zahra

H

,

Frappier

D

,

Burkett

K

,

Carpenter

TO

,

Anderson

D

,

Garabedian

M

,

Sermet

I

,

Fujiwara

TM

,

Morgan

K

,

Tenenhouse

HS

,

Juppner

H

2006

SLC34A3 mutations in patients with hereditary hypophosphatemic rickets with hypercalciuria predict a key role for the sodium-phosphate cotransporter NaPi-IIc in maintaining phosphate homeostasis.

Am J Hum Genet

78

:

179

192

35.

Hoopes

RR

,

Raja

KM

,

Koich

A

,

Hueber

P

,

Reid

R

,

Knohl

SJ

,

Scheinman

SJ

2004

Evidence for genetic heterogeneity in Dent's disease.

Kidney Int

65

:

1615

1620

36.

Nesterova

G

,

Gahl

WA

1993

Cystinosis

. In: ,

Pagon

R

,

Bird

T

,

Dolan

C

,

Stephens

K

, eds.

GeneReviews

.

Seattle

:

University of Washington

.

37.

Magen

D

,

Berger

L

,

Coady

MJ

,

Ilivitzki

A

,

Militianu

D

,

Tieder

M

,

Selig

S

,

Lapointe

JY

,

Zelikovic

I

,

Skorecki

K

2010

A loss-of-function mutation in NaPi-IIa and renal Fanconi's syndrome.

N Engl J Med

362

:

1102

1109

38.

Nafidi

O

,

Lapointe

RW

,

Lepage

R

,

Kumar

R

,

D'Amour

P

2009

Mechanisms of renal phosphate loss in liver resection-associated hypophosphatemia.

Ann Surg

249

:

824

827

39.

Mayne

PD

,

Kovar

IZ

1991

Calcium and phosphorus metabolism in the premature infant.

Ann Clin Biochem

28

:

131

142

40.

Laaban

JP

,

Grateau

G

,

Psychoyos

I

,

Laromiguière

M

,

Vuong

TK

,

Rochemaure

J

1989

Hypophosphatemia induced by mechanical ventilation in patients with chronic obstructive pulmonary disease.

Crit Care Med

17

:

1115

1120

41.

Roestel

C

,

Hoeping

W

,

Deckert

J

2004

Hypophosphatemia in panic disorder.

Am J Psychiatry

161

:

1499

1500

42.

Halevy

J

,

Bulvik

S

1988

Severe hypophosphatemia in hospitalized patients.

Arch Intern Med

148

:

153

155

43.

Schouten

BJ

,

Doogue

MP

,

Soule

SG

,

Hunt

PJ

2009

Iron polymaltose-induced FGF23 elevation complicated by hypophosphataemic osteomalacia.

Ann Clin Biochem

46

:

167

169

44.

Schouten

BJ

,

Hunt

PJ

,

Livesey

JH

,

Frampton

CM

,

Soule

SG

2009

FGF23 elevation and hypophosphatemia after intravenous iron polymaltose: a prospective study.

J Clin Endocrinol Metab

94

:

2332

2337

45.

Shimizu

Y

,

Tada

Y

,

Yamauchi

M

,

Okamoto

T

,

Suzuki

H

,

Ito

N

,

Fukumoto

S

,

Sugimoto

T

,

Fujita

T

2009

Hypophosphatemia induced by intravenous administration of saccharated ferric oxide: another form of FGF23-related hypophosphatemia.

Bone

45

:

814

816

46.

Hothi

DK

,

Harvey

E

,

Piva

E

,

Keating

L

,

Secker

D

,

Geary

DF

2006

Calcium and phosphate balance in adolescents on home nocturnal haemodialysis.

Pediatr Nephrol

21

:

835

841

47.

Walton

RJ

,

Bijvoet

OLM

1975

Nomogram for derivation of renal threshold phosphate concentration.

Lancet

306

:

309

310

48.

Carpenter

TO

,

Mitnick

MA

,

Ellison

A

,

Smith

C

,

Insogna

KL

1994

Nocturnal hyperparathyroidism: a frequent feature of X-linked hypophosphatemia.

J Clin Endocrinol Metab

78

:

1378

1383

49.

Rivkees

SA

,

el-Hajj-Fuleihan

G

,

Brown

EM

,

Crawford

JD

1992

Tertiary hyperparathyroidism during high phosphate therapy of familial hypophosphatemic rickets.

J Clin Endocrinol Metab

75

:

1514

1518

50.

Woo

VL

,

Landesberg

R

,

Imel

EA

,

Singer

SR

,

Folpe

AL

,

Econs

MJ

,

Kim

T

,

Harik

LR

,

Jacobs

TP

2009

Phosphaturic mesenchymal tumor, mixed connective tissue variant, of the mandible: report of a case and review of the literature.

Oral Surg Oral Med Oral Pathol Oral Radiol Endod

108

:

925

932

51.

Moran

M

,

Paul

A

2002

Octreotide scanning in the detection of a mesenchymal tumour in the pubic symphysis causing hypophosphataemic osteomalacia.

Int Orthop

26

:

61

62

52.

Dupond

JL

,

Mahammedi

H

,

Prié

D

,

Collin

F

,

Gil

H

,

Blagosklonov

O

,

Ricbourg

B

,

Meaux-Ruault

N

,

Kantelip

B

2005

Oncogenic osteomalacia: diagnostic importance of fibroblast growth factor 23 and F-18 fluorodeoxyglucose PET/CT scan for the diagnosis and follow-up in one case.

Bone

36

:

375

378

53.

Hesse

E

,

Moessinger

E

,

Rosenthal

H

,

Laenger

F

,

Brabant

G

,

Petrich

T

,

Gratz

KF

,

Bastian

L

2007

Oncogenic osteomalacia: exact tumor localization by co-registration of positron emission and computed tomography.

J Bone Miner Res

22

:

158

162

54.

Shulman

DI

,

Hahn

G

,

Benator

R

,

Washington

K

,

White

KE

,

Farber

J

,

Econs

MJ

2004

Tumor-induced rickets: usefulness of MR gradient echo recall imaging for tumor localization.

J Pediatr

144

:

381

385

55.

Ward

LM

,

Rauch

F

,

White

KE

,

Filler

G

,

Matzinger

MA

,

Letts

M

,

Travers

R

,

Econs

MJ

,

Glorieux

FH

2004

Resolution of severe, adolescent-onset hypophosphatemic rickets following resection of an FGF-23-producing tumour of the distal ulna.

Bone

34

:

905

911

56.

David

K

,

Revesz

T

,

Kratimenos

G

,

Krausz

T

,

Crockard

HA

1996

Oncogenic osteomalacia associated with a meningeal phosphaturic mesenchymal tumor. Case report.

J Neurosurg

84

:

288

292

57.

Nasu

T

,

Kurisu

S

,

Matsuno

S

,

Tatsumi

K

,

Kakimoto

T

,

Kobayashi

M

,

Nakano

Y

,

Wakasaki

H

,

Furuta

H

,

Nishi

M

,

Sasaki

H

,

Suzuki

H

,

Ito

N

,

Fukumoto

S

,

Nanjo

K

2008

Tumor-induced hypophosphatemic osteomalacia diagnosed by the combinatory procedures of magnetic resonance imaging and venous sampling for FGF23.

Intern Med

47

:

957

961

58.

Westerberg

PA

,

Olauson

H

,

Toss

G

,

Wikström

B

,

Morales

O

,

Linde

T

,

Jonsson

K

,

Ljunggren

O

,

Larsson

TE

2008

Preoperative tumor localization by means of venous sampling for fibroblast growth factor-23 in a patient with tumor-induced osteomalacia.

Endocr Pract

14

:

362

367

59.

Ito

N

,

Shimizu

Y

,

Suzuki

H

,

Saito

T

,

Okamoto

T

,

Hori

M

,

Akahane

M

,

Fukumoto

S

,

Fujita

T

2010

Clinical utility of systemic venous sampling of FGF23 for identifying tumours responsible for tumour-induced osteomalacia.

J Intern Med

268

:

390

394

60.

Takeuchi

Y

,

Suzuki

H

,

Ogura

S

,

Imai

R

,

Yamazaki

Y

,

Yamashita

T

,

Miyamoto

Y

,

Okazaki

H

,

Nakamura

K

,

Nakahara

K

,

Fukumoto

S

,

Fujita

T

2004

Venous sampling for fibroblast growth factor-23 confirms preoperative diagnosis of tumor-induced osteomalacia.

J Clin Endocrinol Metab

89

:

3979

3982

61.

Andreopoulou

P

,

Dumitrescu

CE

,

Kelly

MH

,

Brillante

BA

,

Cutler Peck

CM

,

Wodajo

FM

,

Chang

R

,

Collins

MT

2011

Selective venous catheterization for the localization of phosphaturic mesenchymal tumors.

J Bone Miner Res

26

:

1295

1302

62.

Reid

IR

,

Murphy

WA

,

Hardy

DC

,

Teitelbaum

SL

,

Bergfeld

MA

,

Whyte

MP

1991

X-linked hypophosphatemia: skeletal mass in adults assessed by histomorphometry, computed tomography, and absorptiometry.

Am J Med

90

:

63

69

63.

Camp

MA

,

Allon

M

1990

Severe hypophosphatemia in hospitalized patients.

Miner Electrolyte Metab

16

:

365

368

64.

Zazzo

JF

,

Troché

G

,

Ruel

P

,

Maintenant

J

1995

High incidence of hypophosphatemia in surgical intensive care patients: efficacy of phosphorus therapy on myocardial function.

Intensive Care Med

21

:

826

831

65.

Taylor

BE

,

Huey

WY

,

Buchman

TG

,

Boyle

WA

,

Coopersmith

CM

2004

Treatment of hypophosphatemia using a protocol based on patient weight and serum phosphorus level in a surgical intensive care unit.

J Am Coll Surg

198

:

198

204

66.

Clark

CL

,

Sacks

GS

,

Dickerson

RN

,

Kudsk

KA

,

Brown

RO

1995

Treatment of hypophosphatemia in patients receiving specialized nutrition support using a graduated dosing scheme: results from a prospective clinical trial.

Crit Care Med

23

:

1504

1511

67.

Vannatta

JB

,

Andress

DL

,

Whang

R

,

Papper

S

1983

High-dose intravenous phosphorus therapy for severe complicated hypophosphatemia.

South Med J

76

:

1424

1426

68.

Rosen

GH

,

Boullata

JI

,

O'Rangers

EA

,

Enow

NB

,

Shin

B

1995

Intravenous phosphate repletion regimen for critically ill patients with moderate hypophosphatemia.

Crit Care Med

23

:

1204

1210

69.

Tenenhouse

HS

,

Econs

MJ

2001

Mendelian hypophosphatemias

. In: ,

Scriver

CR

,

Beaudet

AL

,

Valle

D

,

Sly

WS

,

Vogelstein

B

,

Childs

B

,

Kinzler

KW

, eds.

The metabolic and molecular bases of inherited disease

.

New York

:

McGraw-Hill Professional

;

5039

5067

70.

Carpenter

TO

,

Imel

EA

,

Holm

IA

,

Jan de Beur

SM

,

Insogna

KL

2011

A clinician's guide to X-linked hypophosphatemia.

J Bone Miner Res

26

:

1381

1388

71.

Harrell

RM

,

Lyles

KW

,

Harrelson

JM

,

Friedman

NE

,

Drezner

MK

1985

Healing of bone disease in X-linked hypophosphatemic rickets/osteomalacia. Induction and maintenance with phosphorus and calcitriol.

J Clin Invest

75

:

1858

1868

72.

Glorieux

FH

,

Marie

PJ

,

Pettifor

JM

,

Delvin

EE

1980

Bone response to phosphate salts, ergocalciferol, and calcitriol in hypophosphatemic vitamin D-resistant rickets.

N Engl J Med

303

:

1023

1031

73.

Lyles

KW

,

Harrelson

JM

,

Drezner

MK

1982

The efficacy of vitamin D2 and oral phosphorus therapy in X-linked hypophosphatemic rickets and osteomalacia.

J Clin Endocrinol Metab

54

:

307

315

74.

Balsan

S

,

Tieder

M

1990

Linear growth in patients with hypophosphatemic vitamin D-resistant rickets: influence of treatment regimen and parental height.

J Pediatr

116

:

365

371

75.

Verge

CF

,

Lam

A

,

Simpson

JM

,

Cowell

CT

,

Howard

NJ

,

Silink

M

1991

Effects of therapy in X-linked hypophosphatemic rickets.

N Engl J Med

325

:

1843

1848

76.

Taylor

A

,

Sherman

NH

,

Norman

ME

1995

Nephrocalcinosis in X-linked hypophosphatemia: effect of treatment versus disease.

Pediatr Nephrol

9

:

173

175

77.

Imel

EA

,

DiMeglio

LA

,

Hui

SL

,

Carpenter

TO

,

Econs

MJ

2010

Treatment of X-linked hypophosphatemia with calcitriol and phosphate increases circulating fibroblast growth factor 23 concentrations.

J Clin Endocrinol Metab

95

:

1846

1850

78.

Girgis

CM

,

Wong

T

,

Ngu

MC

,

Emmett

L

,

Archer

KA

,

Chen

RC

,

Seibel

MJ

2011

Hypophosphataemic osteomalacia in patients on adefovir dipivoxil.

J Clin Gastroenterol

45

:

468

473

79.

François

H

,

Coppo

P

,

Hayman

JP

,

Fouqueray

B

,

Mougenot

B

,

Ronco

P

2008

Partial Fanconi syndrome induced by imatinib therapy: a novel cause of urinary phosphate loss.

Am J Kidney Dis

51

:

298

301

80.

Saidenberg-Kermanac'h

N

,

Souabni

L

,

Prendki

V

,

Prie

D

,

Boissier

MC

2011

Normal plasma FGF23 levels kinetic in tenofovir-related hypophosphatemic osteomalacia in an HIV-infected patient with von Recklinghausen disease.

Joint Bone Spine

78

:

306

308

81.

Bellini

E

,

Pia

A

,

Brizzi

MP

,

Tampellini

M

,

Torta

M

,

Terzolo

M

,

Dogliotti

L

,

Berruti

A

2011

Sorafenib may induce hypophosphatemia through a fibroblast growth factor-23 (FGF23)-independent mechanism.

Ann Oncol

22

:

988

990

82.

Miller

DW

,

Slovis

CM

2000

Hypophosphatemia in the emergency department therapeutics.

Am J Emerg Med

18

:

457

461

83.

Stanga

Z

,

Brunner

A

,

Leuenberger

M

,

Grimble

RF

,

Shenkin

A

,

Allison

SP

,

Lobo

DN

2008

Nutrition in clinical practice—the refeeding syndrome: illustrative cases and guidelines for prevention and treatment.

Eur J Clin Nutr

62

:

687

694

Copyright © 2012 by The Endocrine Society

Copyright © 2012 by The Endocrine Society

Citations

Views

Altmetric

Email alerts

More on this topic

Related articles in PubMed

Citing articles via

  • Latest

  • Most Read

  • Most Cited

More from Oxford Academic

Which of the following conditions is associated with hypophosphatemia?

Treatment. Hypophosphatemia affects the entire body, even on an intracellular level. If gone untreated, it can result in: Bone diseases like rickets, osteopenia, osteoporosis, and osteomalacia.

What causes the clinical manifestations of severe symptomatic hypophosphatemia?

The major conditions associated with symptomatic hypophosphatemia are chronic alcoholism, intravenous hyperalimentation without phosphate supplementation, urinary phosphate-wasting syndromes (such as Fanconi syndrome or tumor-induced osteomalacia), and the chronic ingestion of antacids or other phosphate binders.

Which of the following is the most important assessment parameter the nurse should consider when assessing a fluid and electrolyte imbalance?

Cardiac rate and rhythm are the most important physical assessment parameter to measure. Skin turgor, intake and output are physical assessment parameters a nurse would consider when assessing fluid and electrolyte imbalance, but choice d is the most important.

Which patient is at most risk for hypomagnesemia?

Medical conditions putting persons at high risk for hypomagnesemia are alcoholism, congestive heart failure, diabetes, chronic diarrhea, hypokalemia, hypocalcemia, and malnutrition (strength of recommendation: C, based on expert opinion, physiology, and case series).

Toplist

Neuester Beitrag

Stichworte