HIPER_HIPOPARATIR_NEJM

download HIPER_HIPOPARATIR_NEJM

of 13

Transcript of HIPER_HIPOPARATIR_NEJM

  • 8/12/2019 HIPER_HIPOPARATIR_NEJM

    1/13

    MEDICAL PROGRESS

    Vol ume 343 Nu mb er 25 1863

    Medical Progress

    H

    YPERPARATHYROID

    AND

    H

    YPOPARATHYROID

    D

    ISORDERS

    S

    TEPHEN

    J. M

    ARX

    , M.D.

    From the Metabolic Diseases Branch, National Institute of Diabetes andDigestive and Kidney Diseases, Bethesda, Md. Address reprint requests toDr. Marx at the National Institute of Diabetes and Digestive and KidneyDiseases, Bldg. 10, Rm. 9C-101, National Institutes of Health, 9000Rockville Pike, Bethesda, MD 20892-1802, or at [email protected].

    2000, Massachusetts Medical Society.

    HE four parathyroid glands, through the secre-tion of parathyroid hormone, regulate serumcalcium concentrations and bone metabolism.

    1

    In turn, serum calcium concentrations regulate par-athyroid hormone secretion; high concentrations in-hibit secretion by the parathyroid glands of parathy-roid hormone and low concentrations stimulate it.

    2

    Low or falling serum calcium concentrations act with-in seconds to stimulate parathyroid hormone secre-tion, initiated by means of a calcium-sensing receptoron the surface of the parathyroid cells.

    2

    This receptor

    is a heptahelical molecule, like the receptors for light,odorants, catecholamines, and many peptide hor-mones.

    3

    Parathyroid hormone secretion is 50 per-cent of the maximal level at a serum ionized calciumconcentration of 4 mg per deciliter (1 mmol per li-ter); this is considered the calcium set point for par-athyroid hormone secretion. A slower regulation ofparathyroid hormone secretion occurs over a periodof hours as a result of cellular changes in parathyroidhormone messenger RNA (mRNA). Vitamin D andits metabolites 25-hydroxyvitamin D and 1,25-dihy-droxyvitamin D, acting through vitamin D receptors,decrease the level of parathyroid hormone mRNA,

    4

    and hypocalcemia increases the level of that mRNA.

    5,6

    The slowest regulation of parathyroid hormone se-cretion occurs over days or even months and reflectschanges in the growth of the parathyroid glands.Metabolites of vitamin D directly inhibit the mass ofparathyroid cells

    7

    ; hypocalcemia stimulates the growthof parathyroid cells independently of the contraryaction of vitamin D metabolites.

    8,9

    Disruptions inthese processes cause hyperparathyroidism or hypo-parathyroidism.

    STRUCTURE AND ACTIONS OF

    PARATHYROID HORMONE

    Parathyroid hormone is stored and secreted main-ly as an 84-amino-acid peptide.

    1

    A synthetic amino-

    terminal fragment, parathyroid hormone (134), isfully active; modifications at the amino terminal, par-ticularly at the first two residues, can abolish its bio-

    T

    logic activity.

    10

    The effects of parathyroid hormone onmineral metabolism are initiated by the binding ofparathyroid hormone to the type 1 parathyroid hor-mone receptor in the target tissues.

    11

    Parathyroid hor-mone thereby regulates large calcium fluxes acrossbone, kidneys, and intestines

    1

    (Fig. 1). Another par-

    athyroid hormone receptor (type 2) has been foundin the brain and the intestines. Its main ligand is a pep-tide different from parathyroid hormone

    12

    ; the func-tions of this receptor are not known.

    Parathyroid hormonerelated peptide is a distanthomologue of parathyroid hormone and is not a truehormone. It is synthesized in cartilage and in manymore tissues than is parathyroid hormone, and its se-cretion is not regulated by serum calcium.

    13

    Its localrelease activates the type 1 parathyroid hormone re-ceptor, and its affinity for this receptor is similar tothat of parathyroid hormone (Fig. 1).

    MEASUREMENT OF PARATHYROID

    HORMONE IN SERUM

    Measurements of serum calcium, parathyroid hor-mone, 25-hydroxyvitamin D, and 1,25-dihydroxyvi-tamin D are used regularly in the diagnosis and treat-ment of hyperparathyroidism and hypoparathyroidism;only the measurement of serum parathyroid hormoneis covered here. Serum calcium should usually bemeasured at the same time as serum parathyroid hor-mone; since the ionized fraction of serum calcium isthe biologically active form, it is a more useful indexof hyperparathyroidism and hypoparathyroidism thanare other indexes of calcium in serum. It is thereforethe preferred form of serum calcium to measure.

    Current assays for serum parathyroid hormone aretwo-site assays designed to detect both amino-termi-nal and carboxy-terminal epitopes of the peptide.

    14,15

    The better assays are those that are well standardized,do not cross-react with parathyroid hormonerelat-ed peptide, and are sufficiently sensitive that normal

    values can be distinguished from subnormal values(Fig. 2). Parathyroid hormone molecules that are re-active in these two-site immunoassays are consideredintact, but some have no bioactivity.

    14-17

    For exam-ple, a loss of only six amino acids to yield parathyroidhormone (784) eliminates all bioactivity but doesnot affect the immunoreactivity measured in most orall of these assays.

    10

    In fact, about half of the para-thyroid hormone detected with these assays in theserum of patients with chronic renal disease is bio-logically inactive.

    16,17

    Measurements of parathyroid hormone can helpcharacterize parathyroid tumors. Parathyroid hormonecan be measured in fluid obtained from a lesion byfine-needle aspiration (usually guided ultrasonograph-ically) or in serum from the veins of the neck andmediastinum, catheterized selectively.

    18

    Serum test re-sults that can be obtained in 10 to 15 minutes allowphysicians to assess the completeness of the removal

  • 8/12/2019 HIPER_HIPOPARATIR_NEJM

    2/13

    1864

    December 21, 2000

    The New England Journal of Medicine

    of hyperfunctioning parathyroid tissue during the

    operation.

    19

    PRIMARY HYPERPARATHYROIDISM

    Most patients with primary hyperparathyroidismhave high serum parathyroid hormone concentrations.Most also have high serum calcium concentrations,and even more have high serum ionized calcium con-centrations. The most important diagnostic tests forthis disorder are thus measurements of serum para-thyroid hormone and ionized calcium (Fig. 2).

    The Parathyroid Gland in Primary Hyperparathyroidism

    Solitary parathyroid adenomas are monoclonal oroligoclonal tumors.

    20

    Similarly, in multiglandular hy-perparathyroidism, most of the parathyroid tumors aremonoclonal or oligoclonal,

    21

    reflecting overgrowthfrom somatic or germ-line mutations in parathyroid-tumor precursor cells. The underlying genes that de-

    velop mutations in hyperparathyroidism have beenidentified only in a minority of tumors. They help topinpoint the molecular pathway of oncogenesis andthus help to determine possible targets for treatment

    Figure 1.

    The Parathyroid Axis.

    The synthesis of parathyroid hormone (PTH) and parathyroid hormonerelated peptide (PTHrP) is shown on the left, and their targetsites of action are shown on the right. Both act by means of the same receptor (also termed the type 1 PTH receptor). Negative

    feedback of 1,25-dihydroxyvitamin D is not shown. See the text for further descriptions. An excess or deficiency of parathyroid hor-mone may be treated either at the level of parathyroid hormone release (and the parathyroid hormone receptors) or at selectedsites distal to the parathyroid hormone receptors. Blue arrows indicate extracellular calcium flow.

    Calcium-

    sensing

    receptor

    Calcium-

    sensing

    receptor

    Renal

    tubule

    Extracellular ionized

    calcium

    Bone

    Parathyroid cell

    PTH

    Endocrine

    mechanism

    PTHrP

    PTHrP

    Autocrineparacrine

    mechanism

    PTH receptor

    PTH

    receptor

    1,25(OH)2D

    Duodenal

    lumen

    Cartilage and PTHrP

    target cells in many

    other tissues

    Blood and other

    extracellular fluid

    Ca2+

    Ca2+

    Ca2+

  • 8/12/2019 HIPER_HIPOPARATIR_NEJM

    3/13

    MEDICAL PROGRESS

    Vol ume 343 Nu mb er 25

    1865

    (Fig. 3).

    22-27

    As in other tumors, it is likely that twoor more genes have mutated in parathyroid adenomas,reflecting a stepwise development of the adenoma.

    28-31

    Many of the known and unknown genes that havemutated in parathyroid tumors are probably tumor-suppressor genes; that is to say, they contribute to theformation of the tumor through a sequential inacti-

    vation of both copies of the gene.

    31-34

    The multipleendocrine neoplasia type 1 gene (

    MEN1

    ) is a tumor-suppressor gene and the known gene that most of-ten has somatic mutations in both copies in parathy-roid adenomas (in 20 percent of cases).

    35

    Since thecalcium-sensing receptor and the vitamin D receptoralso mediate the inhibition of parathyroid-gland func-tion, it is noteworthy that no inactivating mutationof either gene has been identified in parathyroid ad-enomas.

    35-38

    A small minority of parathyroid adenomas have

    activating mutations of the cyclin D1 gene(

    CCND1

    ).

    20,24,39

    These mutations result in the over-expression of the protein cyclin D1, but cyclin D1overexpression is even more common in parathyroidadenomas without cyclin D1 mutations.

    40

    The abnormal parathyroid cells in primary (andsecondary) hyperparathyroidism have deficient sen-sitivity to inhibition by calcium; this may result in partfrom a deficiency of calcium-sensing receptors on par-athyroid cells.

    41

    Deficiency of these receptors is prob-ably a consequence and not a cause of neoplastic pri-mary hyperparathyroidism.

    Categories and Causes of Sporadic PrimaryHyperparathyroidism

    Solitary parathyroid adenomas account for 85 per-cent of cases of primary hyperparathyroidism; hyper-function in multiple parathyroid glands (a broad cat-

    Figure 2.

    Serum Calcium and Parathyroid Hormone Concentrations in Patients with Hypercalcemia andHypocalcemia Due to Various Causes.

    The diagnosis of a serious mineral disorder is usually clear, as illustrated by the nonoverlapping do-mains in the figure, but in the early stages of these disorders, the values for either serum calcium or

    parathyroid hormone may overlap with the normal ranges. The following diagnoses are not shown:familial hypocalciuric hypercalcemia (midpoint of the range for serum calcium, 11.5 mg per deciliter,and for serum parathyroid hormone, 30 pg per milliliter); neonatal severe primary hyperparathyroid-

    ism (midpoint for serum calcium, 18 mg per deciliter, and for serum parathyroid hormone, 500 pg permilliliter); hypercalciuric hypocalcemia (midpoint for serum calcium, 7 mg per deciliter, and for serumparathyroid hormone, 10 pg per milliliter); tertiary uremic hyperparathyroidism (midpoint for serum

    calcium, 11 mg per deciliter, and for serum parathyroid hormone, 2000 pg per milliliter); tertiary hy-perparathyroidism after renal transplantation that corrected uremia (midpoint for serum calcium, 12mg per deciliter, and for serum parathyroid hormone, 200 pg per milliliter); and adynamic bone dis-

    ease with uremia (midpoint for serum calcium, 9 mg per deciliter, and for serum parathyroid hormone,

    50 pg per milliliter). To convert values for serum calcium to millimoles per liter, multiply by 0.25, andto convert values for serum parathyroid hormone to picomoles per liter, multiply by 0.11.

    10,000

    6

    1

    Primaryhyperpara-thyroidism

    Primaryhyperparathyroidism

    Tumorhypercalcemia

    Normal

    Lower limitof detection

    Uremichyperpara-

    thyroidism

    10

    100

    1,000

    7 8 9 10 11 12 13 14 15

    Serum Calcium (mg/dl)

    SerumP

    arathyroidHormon

    e(pg/ml)

  • 8/12/2019 HIPER_HIPOPARATIR_NEJM

    4/13

    1866

    December 21, 2000

    The New England Journal of Medicine

    egory that includes hyperplasia, multiple adenomas,and polyclonal hyperfunction) occurs in most of theremainder; and a few patients (less than 1 percent)have parathyroid carcinoma. About 75 percent of pa-tients with sporadic primary hyperparathyroidism are

    women; the average age at diagnosis is 55 years. The

    annual incidence of primary hyperparathyroidismamong postmenopausal women in Olmsted County,Minnesota, peaked at 112 per 100,000 in 1974; thehigh number of diagnoses in that year is attributableto the introduction of screening measurements of se-rum calcium. The annual incidence then fell markedly

    Figure 3.

    Protein That Causes Parathyroid-Gland Hyperfunction When Mutated.

    Mutation may occur through inheritance (germ-line mutation) or postnatally (somatic mutation) in abnormal parathyroid tissue. In-activating mutations characterize tumor-suppressorencoded proteins (menin [the product of the MEN1

    tumor-suppressor gene],p53, and retinoblastoma protein [pRb]), which are shown in red. Similarly, the calcium-sensing receptor is a growth suppressor

    (shown in red). Activating mutations characterize proto-oncoproteins (cyclin D1 and RET) and are shown in yellow. Menin binds toJunD, a transcription factor, which dimerizes (connects) with another member of the JunFos family of transcription factors; meninthereby inhibits transcriptional activation by JunD.

    22

    The p53 protein binds to DNA through a specific DNA response element.

    23

    Cyclin D1 is a cell-cycle regulator that activates the catalytic units of cyclin-dependent kinases (CDK) 4 and 6.

    24

    One substrate forphosphorylation (P) and thus blockade by CDK 4 or 6 is pRb,

    25

    which binds to the transcription factor E2F as well as to several othertranscription factors.

    25

    Black T bars indicate binding to a specific sequence of DNA. Calcium ions probably bind to the calcium-

    sensing receptor in the plasma membrane, which transmits information on extracellular calcium to an unidentified guanine-nucle-otidebinding protein (G

    ?

    ) in the cytoplasm.

    26

    The RET

    -encoded tyrosine kinase in the plasma membrane (RET [yellow]) is a dimerthat phosphorylates Src-homology collagen (SHC) and other substrates. RET is regulated by an extracellular RET receptor attached

    to the membrane by its glycosylphosphatidylinositol anchor (RR-GPI [turquoise]).

    27

    There are at least four extracellular RET recep-tors, each with different extracellular protein ligands (RRL). The full mechanisms by which any of these mutant proteins contributeto tumor formation are not known. Arrows show the flow of a molecule to or away from the plasma membrane.

    JunDMenin

    PTH

    CDK 4 or 6

    CyclinD1

    pRb

    SHC

    RET

    CDK 4 or 6

    CyclinD1

    pRb

    DNA

    P E2FE2F

    p53

    SHC

    RET

    RR-GPI

    RRL

    Calcium-sensing

    receptor

    Secretory

    granule

    Ca2+

    G?

    Cytoplasm

    Plasma membrane

    Nucleus

  • 8/12/2019 HIPER_HIPOPARATIR_NEJM

    5/13

  • 8/12/2019 HIPER_HIPOPARATIR_NEJM

    6/13

    1868

    December 21, 2000

    The New England Journal of Medicine

    Multiple Endocrine Neoplasia Type 2a

    Multiple endocrine neoplasia type 2a is character-ized primarily by medullary thyroid carcinoma andpheochromocytoma.

    27

    Primary hyperparathyroidismcan occur by the age of 70 in up to 70 percent ofpatients and is usually mild.

    51

    Multiple endocrine neo-plasia type 2a is caused by an activating mutation ofthe RET

    proto-oncogene and is inherited as an au-tosomal dominant trait.

    27

    HyperparathyroidismJaw Tumor Syndrome

    The hyperparathyroidismjaw tumor syndrome israre and is characterized by hyperparathyroidism, ce-mento-ossifying fibromas of the jaw, renal cysts,

    Wilms tumor, and renal hamartomas.47,52,53 By the ageof 40, about 80 percent of patients with this syn-drome have hyperparathyroidism, and about 10 per-cent of those have a parathyroid carcinoma. Often,at the first presentation of hyperparathyroidism, onlyone parathyroid adenoma is present, but multiple ad-

    enomas can occur either simultaneously or at differ-ent times. The disorder is caused by a mutation inan unknown gene on chromosome 1q2452 and is in-herited as an autosomal dominant trait.

    Manifestations of Primary Hyperparathyroidism

    The parathyroids are small endocrine glands, andincreases in their size or enhancements of their func-tion have no effect on neighboring tissues. Instead,the effects of an excessive secretion of parathyroid hor-mone are manifested chemically as abnormal fluxes ofcalcium and phosphate in bone, in the kidneys, andin the gastrointestinal tract (Fig. 1). The main resultsare hypercalcemia, hypercalciuria, and increased rates

    of bone turnover.Primary hyperparathyroidism is usually first sus-

    pected when a patient is found on biochemical screen-ing to have hypercalcemia; less often it is suspectedbecause nephrolithiasis or osteopenia is present. Theanticalciuric effect of thiazide drugs can raise serumcalcium concentrations slightly, thereby uncoveringoccult hyperparathyroidism. With the current restric-tions on reimbursement for biochemical screening,the proportion of newly diagnosed cases of hyper-parathyroidism that are asymptomatic should decrease.

    Currently, most patients in whom hyperparathy-roidism is diagnosed at first appear to be asympto-matic,54,55 but up to half of them have subtle neuro-

    behavioral symptoms such as fatigue and weakness.56,57In many of these patients, the fact that fatigue or

    weakness is a symptom of hyperparathyroidism be-comes clear only after a successful parathyroidectomy,

    when the symptom resolves. About 20 percent ofpatients with hyperparathyroidism have nephrolithi-asis.55 Primary hyperparathyroidism can cause cardi-ac calcifications and left ventricular hypertrophy; thelatter can occur in the absence of hypertension andcan be partially reversed after parathyroidectomy.58

    There is some controversy about whether any of thesechanges decrease life expectancy. A recent population-based study found that there was no excess mortalityamong all patients with hyperparathyroidism, butthere was excess mortality among the patients in thehighest quartile for serum calcium concentrations.59

    Effects on Bone

    Parathyroid hormone increases the rate of boneturnover, and its effects on bone may be catabolic oranabolic, depending on the age of the patient, theskeletal site, and the pattern of serum concentrationsof the hormone over time.60,61In general, persistent-ly high serum parathyroid hormone concentrationshave catabolic effects on bone, whereas intermittentmild increases have anabolic effects.

    On balance, the effects of mild primary hyperpara-thyroidism on bone seem to be slightly anabolic.62

    However, the disorder can cause a demineralization

    of bone, distributed variably between cortical sites (i.e.,mainly long bones) and trabecular sites (i.e., mainlyvertebrae).63,64Approximately one in four patients hasosteopenia (a z score lower than 2; z refers to thenumber of standard deviations from an age- and sex-matched mean) in cortical or trabecular bone.63,64

    Overall, the risk of bone fractures in patients withmild hyperparathyroidism is similar to that in matchednormal subjects (one new fracture per decade); still,the presence of hyperparathyroidism significantly in-creased the risk of fracture in several bones, particu-larly the vertebrae, in a population-based, controlledstudy.65Successful parathyroidectomy is followed byan increase in bone mass over a period of 6 to 12

    months,55,63,66

    with continued increases for up to 10years after surgery.55

    Natural History and Treatment of PrimaryHyperparathyroidism

    In most patients, primary hyperparathyroidism pro-gresses slowly, if at all. Among asymptomatic patients,only about 25 percent have progressive disease, whichis usually manifested as a decrease in bone mass dur-ing a 10-year period of follow-up.55Thus, there hasbeen some controversy regarding the indications forsurgery, the only effective treatment. A consensus con-ference of the National Institutes of Health conclud-ed in 1990 that surgery was not routinely needed in

    asymptomatic patients 50 years old or older whohad a serum calcium concentration 1.0 to 1.6 mg perdeciliter (0.25 to 0.40 mmol per liter) above the up-per limit of normal, a level of urinary calcium excre-tion of less than 400 mg (100 mmol) per day, a cre-atinine clearance of at least 70 percent of normal, ora z score higher than 2 for bone mass.67These arestill reasonable guidelines, but surgery may be rec-ommended for many of these patients because of theevidence that it ameliorates neurobehavioral symp-

  • 8/12/2019 HIPER_HIPOPARATIR_NEJM

    7/13

    MEDICAL PROGRESS

    Vol ume 343 Nu mb er 25 1869

    toms that may be hard to detect.56,57Surgery is notonly an effective but also a safe treatment for primaryhyperparathyroidism, even in patients who are morethan 70 years old.68

    Several methods for characterizing overactive par-athyroid glands are available, including ultrasonog-

    raphy and imaging with technetium-99m sestamibibefore or during surgery (Fig. 4), but these are notused routinely.69-71Rapid assays for measuring serumparathyroid hormone during surgery are also available,as discussed above. So-called minimally invasive sur-gical methods have become possible because imagingcan be used to detect parathyroid adenomas and cansometimes be coupled with a rapid assay of parathy-roid hormone during surgery.72Such approaches candecrease the duration of the surgery, but the rate ofsuccess may not match that of standard parathyroid-ectomy.73,74When patients require repeated operation,every effort should be made to identify abnormaltissue preoperatively, and intraoperative testing is of-

    ten included as well.18,19Patients who do not undergo surgery should be

    evaluated clinically, and serum calcium, creatinine,and parathyroid hormone should be measured at6-to-12-month intervals, and cortical and trabecularbone density at 12-month intervals. Such patientsshould be advised to avoid dehydration and to keeptheir calcium intake at or below 1000 mg per day.

    Some patients with more severe primary hyper-parathyroidism may not undergo surgery because ofcontraindications or because they decline the proce-dure; in others, surgery may have been unsuccessful.For these patients, several treatments directed at thetarget tissues of parathyroid hormone action are avail-

    able (Table 2).75Bisphosphonates such as alendronateand clodronate inhibit bone resorption; however, theymay be less effective in patients with hyperparathy-roidism than in those with hypercalcemia from othercauses.76Estrogen increases bone density in postmeno-pausal women with hyperparathyroidism but has littleeffect on serum calcium concentrations.77A calcium-sensingreceptor agonist acts directly on parathyroidcells by way of the calcium-sensing receptor (and isthus calcimimetic) in order to inhibit the secretionof parathyroid hormone78; the further developmentof drugs of this type may provide effective treatmentsfor primary and secondary hyperparathyroidism.79 Pa-tients with primary hyperparathyroidism who havesevere symptomatic hypercalcemia should be treated

    with intravenous saline, a bisphosphonate, furosemide,and in some cases dialysis.75

    Most of these treatments for primary hyperpara-thyroidism change the abnormal transfer of calciumfrom the serum to only one target tissue of parathy-roid hormone action (Table 2 and Fig. 1). Most treat-ments for hypoparathyroidism also affect the transferof calcium along only one of these pathways, albeitin the opposite direction.

    HYPERCALCEMIA MEDIATED BY

    PARATHYROID HORMONERELATED

    PEPTIDE

    Hypercalcemia is sometimes caused by serum fac-tors, which may be released by nonparathyroid tu-mors, whether or not there are skeletal metastases.Most of these tumors are malignant and secrete par-athyroid hormonerelated peptide.13In contrast, hy-

    persecretion of parathyroid hormone by a nonparathy-roid tumor is extremely rare.

    UREMIC HYPERPARATHYROIDISM

    Secondary and Tertiary Hyperparathyroidism

    Hypocalcemia from any cause stimulates parathy-roid hormone secretion, and chronic hypocalcemiaalso stimulates the growth of the parathyroid glands.This secondary hyperparathyroidism usually resolves

    with the treatment of the underlying cause of hypo-calcemia. However, in patients with chronic renal fail-ure, secondary hyperparathyroidism often lasts longerand is more severe than in patients with other hypo-calcemic disorders, such as a deficiency or malabsorp-tion of vitamin D.80Eventually, either before or, moreoften, after renal transplantation, secondary hyperpara-thyroidism can develop into a disorder of oversecre-tion of parathyroid hormone with hypercalcemia (ter-tiary hyperparathyroidism).

    The Parathyroid Gland in Uremia

    Hypercalcemia in patients with uremia who havetertiary hyperparathyroidism might reflect an excessof nearly normal parathyroid cells with a consequent-

    Figure 4.Anterior Planar Image of the Neck and Chest of a Pa-tient with Primary Hyperparathyroidism Obtained with Techne-tium-99m Sestamibi, Showing a Parathyroid Adenoma in the

    Mediastinum.

    The patient had undergone an unsuccessful parathyroid explo-ration. The image shown was obtained two hours after the ad-ministration of 20 mCi of the radionuclide. The lobes of the thy-

    roid and the salivary glands are clearly visible. (Image courtesyof Dr. Clara Chen.)

    Parotid

    Submandibular

    Thyroid

    Mediastinalparathyroid

    Heart

  • 8/12/2019 HIPER_HIPOPARATIR_NEJM

    8/13

    1870 December 21, 2000

    The New England Journal of Medicine

    ly high and nonsuppressible base-line secretion ofparathyroid hormone; in fact, however, it most oftenreflects the secretory dysfunction of autonomouslyfunctioning parathyroid cells.21,80,81Overactive para-thyroid glands that have been removed from patients

    with uremia are usually overgrown with monoclonalor oligoclonal components.22,82-84The cause of pro-gression from early, presumably polyclonal, secondaryhyperplasia of the parathyroid to later monoclonal oroligoclonal tumors is poorly understood.21,80,84Prob-ably, some of the genes that are mutated in the par-athyroid glands of patients with secondary or tertiaryhyperparathyroidism are different from those that aremutated in primary hyperparathyroidism; in partic-ular, MEN1mutations are less frequent in the para-thyroid glands of patients with uremia than in tumorsof patients with sporadic primary hyperparathyroid-ism.82,83

    Bone Disease in Patients with Chronic Renal Diseaseand Hyperparathyroidism

    Bone disease in patients with chronic renal diseaseis caused by both hyperparathyroidism and other fac-tors.85,86Some patients with chronic renal disease havehyperparathyroid uremic bone disease, which is char-acterized by an activation of osteoblasts and osteo-clasts with excess bone resorption. Other patients havean adynamic bone disease or osteomalacia. Adynam-ic bone disease is characterized by low activity of thebone cells, no excess accumulation of matrix, and lit-tle parathyroid hypersecretion.86,87Osteomalacia inrenal failure is characterized by excess accumulationof osteoid and a minimal degree of hyperparathy-roidism and has been associated with the accumula-tion of aluminum in bone. This disorder has becomeless common as a result of the minimization of useof products with high concentrations of aluminum,such as are found in some antacids and dialysis flu-ids.86,87The frequency of hyperparathyroid bone dis-

    ease among patients with uremia is similar to the fre-quency of adynamic bone diseases.86,87

    Uremic hyperparathyroid bone disease is best treat-ed by raising serum calcium concentrations and there-by decreasing parathyroid hormone secretion. Thecause of adynamic bone disease is not known, andthere is no specific treatment.88,89

    Treatment of Hyperparathyroidism in Patientswith Chronic Renal Diseases

    In patients with chronic renal failure, secondary hy-perparathyroidism is caused by hypocalcemia, which,in turn, is caused by hyperphosphatemia and decreasedrenal production of 1,25-dihydroxyvitamin D. Treat-ment is based on raising serum calcium concentra-

    tions by the oral administration of calcium salts; thesesalts also ameliorate hyperphosphatemia by chelatingphosphate in the intestines. Additional measures fortreating hypocalcemia include raising the calciumconcentration in the dialysis fluid and administeringsome form of vitamin D. When treatment is initiatedearly, severe secondary hyperparathyroidism can beprevented or at least delayed. There is some contro-

    versy regarding the most appropriate dosage, type, androute of administration of vitamin D or vitamin Danalogue90,91 and the most appropriate phosphatebinder for these patients.921,25-Dihydroxyvitamin D3(calcitriol) has sometimes been given intravenouslyin pulsed doses in the hope of inhibiting parathyroidhormone secretion without raising serum calcium con-centrations,90,91but calcitriol given orally has similareffects.93

    Severe secondary hyperparathyroidism is an im-portant indication for parathyroidectomy in patients

    with chronic renal disease who cannot be treated ad-equately with the measures described above.94Para-thyroidectomy is also appropriate for some patients

    with tertiary hyperparathyroidism.After renal transplantation, secondary hyperpara-

    *This treatment is not currently available.

    TABLE2.TREATMENTSFORHYPERPARATHYROIDISMANDHYPOPARATHYROIDISM.

    PROCESSAFFECTEDBYTREATMENT

    TREATMENTSFORHYPERPARATHYROIDISM

    TREATMENTSFORHYPOPARATHYROIDISM

    Secretion of parathyroid hormone by parathyroid gland Parathyroidectomy

    Calcium-sensingreceptor agonist*

    Parathyroid autograft

    Activation of receptor for parathyroid hormone Blocker of type 1 receptor* Parathyroid hormone (134)*

    Release of calcium from bone BisphosphonatesEstrogen

    Uptake of calcium from gut Blocker of vitamin D receptor* Vitamin D analogueCalcium salts

    Excretion of calcium in urine Forced natriuresis Thiazide

    Exchange with extracorporeal calcium pool Dialysis Intravenous calcium

  • 8/12/2019 HIPER_HIPOPARATIR_NEJM

    9/13

    MEDICAL PROGRESS

    Vol ume 343 Nu mb er 25 1871

    thyroidism usually regresses over a period of 1 to 10years, but the regression may be incomplete, as re-flected in persistently high serum parathyroid hor-mone concentrations.95About one third of patients

    who receive renal transplants have parathyroid hor-moneinduced hypercalcemia postoperatively that,

    depending on its magnitude and duration, can presenta threat to the renal graft and to other functions.The hypercalcemia usually subsides within months orat most a few years, but 1 to 3 percent of patients re-quire parathyroidectomy an average of three years af-ter renal transplantation because of persistent hyper-calcemia.96

    HYPOPARATHYROIDISM

    Hypoparathyroidism can cause hypocalcemia withconsequent paresthesias, muscle spasms (i.e., tetany),and seizures, especially when it occurs rapidly. In con-trast, chronic hypoparathyroidism generally causes hy-pocalcemia so gradually that the only symptom may

    be visual impairment from cataracts caused by yearsof hypoparathyroidism.

    Diagnosis and Causes

    Like hyperparathyroidism, hypoparathyroidism isdiagnosed on the basis of measurements of serumcalcium and parathyroid hormone (Fig. 2).14,15Thecauses of hypoparathyroidism are diverse, represent-ing disruptions of one or more of the steps in thedevelopment and maintenance of parathyroid hor-mone secretion.

    Damage to the Parathyroid Glands from Surgery

    Injury to or removal of the parathyroid glands dur-

    ing neck surgery is the most common cause of acuteor chronic hypoparathyroidism.

    Developmental Defects in the Parathyroid Glands

    Agenesis of the parathyroid glands occurs in in-fants with the DiGeorge syndrome (and the closelyrelated velocardiofacial syndrome). The manifestationsof these syndromes include incomplete developmentin the branchial arches, resulting in varying degreesof parathyroid and thymic hypoplasia, conotruncalcardiac defects, facial malformations, and learning dis-ability. Both syndromes are associated with rearrange-ments and microdeletions affecting an unknown geneor genes on the short arm of chromosome 22.97Anyresultant defect should be treated, depending on itsseverity.98Isolated agenesis of the parathyroid glandsin one family has been attributed to a recessive de-letion in the gene on chromosome 6 that normallyencodes a transcription factor.99

    Autoimmune Hypoparathyroidism

    Hypoparathyroidism is a prominent componentof autoimmune polyglandular syndrome type 1, alsoknown as autoimmune polyendocrinopathycandi-

    diasisectodermal dystrophy syndrome.100The hypo-parathyroidism, like other manifestations of the syn-drome, occurs during childhood; for this reason andbecause of such associated abnormalities as hypoadre-nalism and intestinal malabsorption, the hypoparathy-roidism may be difficult to treat. The syndrome is in-

    herited as an autosomal recessive trait and is caused bymutations in an autoimmune regulator gene (AIRE)

    with a known sequence but an unknown function.101

    Defects in the Parathyroid Hormone Molecule

    A few cases of familial hypoparathyroidism havebeen described in which the cause was a mutation inthe gene for parathyroid hormone that resulted in thesynthesis of a defective parathyroid hormone mole-cule and undetectable amounts of parathyroid hor-mone in serum.102

    Defective Regulation of Parathyroid Hormone Secretion

    Hypocalcemia and hypercalciuria are the chief fea-

    tures of autosomal dominant hypercalciuric hypocal-cemia, which is caused by activating mutations of theparathyroid and renal calcium-sensing receptor. Thesemutations cause excessive calcium-induced inhibitionof parathyroid hormone secretion. The hypocalcemiais usually mild and asymptomatic. When it is mild,it should be treated cautiously, if at all, because rais-ing serum calcium concentrations further increasesurinary calcium excretion and may cause nephrocal-cinosis.41,103

    TREATMENT OF HYPOPARATHYROIDISM

    Calcium and Vitamin D Analogues

    The main treatments available for patients with

    acute or chronic hypoparathyroidism are calcium salts,vitamin D or vitamin D analogues, and drugs thatincrease renal tubular reabsorption of calcium (i.e., thi-azides) (Table 2). The parathyroid hormonedepend-ent renal production of 1,25-dihydroxyvitamin D isdeficient in all hypoparathyroid states. Therefore, ther-apy with a vitamin D analogue is used to ensure thatthere is a steady serum concentration of an active vi-tamin D analogue. If parathyroid hormone is absentor nonfunctional, its hypocalciuric action cannot oc-cur; therefore, raising the serum calcium concentra-tion may cause hypercalciuria, nephrolithiasis, andrenal damage.

    Patients in whom hypocalcemia develops suddenly for example, after neck surgery are best treated

    with intravenous calcium and with oral or intravenouscalcitriol. Those with chronic hypocalcemia shouldbe treated with oral calcium and either calcitriol or

    vitamin D. Patients in whom the efficacy of treatmentmay vary, such as those with autoimmune polyglan-dular syndrome type 1, are best treated with vitamin Danalogues that have a short half-life. Calcitriol raisesserum calcium concentrations within one or twodays after treatment begins, and its action dissipates

  • 8/12/2019 HIPER_HIPOPARATIR_NEJM

    10/13

    1872 December 21, 2000

    The New England Journal of Medicine

    equally rapidly; the action of vitamin D begins anddissipates over a period of two to four weeks.

    Parathyroid-Tissue Transplantation or ParathyroidHormone

    Transplantation of parathyroid tissue is appealing

    but rarely possible. A parathyroid allograft would re-quire immunosuppression and so would be more dan-gerous than the disease it was meant to treat. Parathy-roid autografts are sometimes placed in the forearmand can consist of either fresh parathyroid tissue orparathyroid tissue removed earlier and cryopreserved.The indication for a parathyroid autograft is a highlikelihood of postoperative hypoparathyroidism. Asmany as half of these grafts fail, and among thosethat survive and function, the potential for late auto-graft-mediated recurrences of hyperparathyroidismis substantial, since the parathyroid tissue used forthe graft was abnormal.84,94

    Patients with hypoparathyroidism have been treated

    successfully with synthetic human parathyroid hor-mone (134) given subcutaneously once daily.104Theincrease in urinary calcium excretion in these patients

    was smaller than that which occurs in patients treat-ed with calcium and calcitriol or vitamin D. Howev-er, synthetic human parathyroid hormone (134) isnot currently available.

    GENETIC DISORDERS OF PARATHYROID

    HORMONE ACTION

    Hereditary defects in parathyroid hormone actionare rare but informative. Each confirms the role of animportant signaling molecule. To some extent, thesestates mimic disorders of parathyroid hormone excess

    or deficiency.

    Defects of the Type 1 Parathyroid Hormone Receptor

    Two defects with opposite effects on the type 1parathyroid hormone receptor have a surprisingly sim-ilar effect on bone growth.11,13

    Jansens Chondrodystrophy

    Jansens chondrodystrophy is characterized by shortlimbs, mild hypercalcemia, and low serum parathyroidhormone concentrations. It is caused by activating mu-tations of the type 1 parathyroid hormone receptor105

    and is inherited as an autosomal dominant trait. It isassociated with increased proliferation and delayedmaturation of chondrocytes, which may weaken thegrowth plates, thereby causing the short limbs.

    Blomstrands Chondrodystrophy

    Blomstrands chondrodystrophy is characterized bygrowth impairment, primarily in the form of shortlimbs. It has been lethal prenatally, and therefore theregulation of serum calcium has not been evaluatedin vivo. It is caused by inactivating mutations of thetype 1 parathyroid hormone receptor106and is inher-

    ited as an autosomal recessive trait. The growth platesshow accelerated calcification and a near-absence ofproliferating chondrocytes.

    Defects of the Stimulatory Guanine-NucleotideBindingProtein

    Parathyroid hormone signaling in cells is mediatedby the type 1 parathyroid hormone receptor, whichthen acts on a stimulatory guanine-nucleotidebind-ing (Gs) protein, which is composed of three sub-units (a, b, and g). The Gsasubunit (encoded by theGNAS1gene) mediates cyclic AMP stimulation byparathyroid hormone and by several other peptidehormones, including thyrotropin.26

    Pseudohypoparathyroidism Type 1a

    Pseudohypoparathyroidism type 1a is characterizedby short stature and other skeletal abnormalities, whichare known collectively as Albrights hereditary osteo-dystrophy, as well as hypocalcemia and high serum

    concentrations of parathyroid hormone. It is causedby inactivating mutations in the asubunit of Gs

    26andis inherited as an autosomal dominant trait. Many pa-tients with pseudohypoparathyroidism type 1a haveresistance not only to parathyroid hormone but alsoto thyrotropin.

    Pseudo-pseudohypoparathyroidism

    Pseudo-pseudohypoparathyroidism occurs in fam-ilies with pseudohypoparathyroidism type 1a. It con-sists of a combination of inactivating mutations ofGNAS1and Albrights osteodystrophy without theresistance to multiple hormones that characterizespseudohypoparathyroidism. The hormone resistance

    is suppressed when the mutated GNAS1gene is in-herited from the father (i.e., paternal imprinting, orsuppression, of the mutant copy occurs in selectedtissues).107,108

    Pseudohypoparathyroidism Type 1b

    Pseudohypoparathyroidism type 1b is characterizedby isolated resistance to parathyroid hormone with-out the accompanying Albrights osteodystrophy. It isassociated with defective methylation within GNAS1,

    which is most likely caused by a mutation in or nearGNAS1.109

    Hypocalcemia in patients with pseudohypoparathy-roidism type 1a or 1b should be treated in the same

    way as it is in patients with true hypoparathyroidism.

    CONCLUSIONS

    Despite a confusing disease nomenclature that isa remnant of past eras, substantial insight has beengained into many disorders of the parathyroid axis.

    With the advent of reliable and specific assays for par-athyroid hormone, the diagnosis of parathyroid dys-function has become much easier. Treatments are gen-erally satisfactory and are logically related to the defects

  • 8/12/2019 HIPER_HIPOPARATIR_NEJM

    11/13

    MEDICAL PROGRESS

    Vol ume 343 Nu mb er 25 1873

    in the parathyroid gland or to their expression in thetarget organs. Controversies persist, however, particu-larly about the treatment of primary hyperparathy-roidism.

    REFERENCES

    1. Jppner H, Gardella TJ, Brown EM, Kronenberg HM, Potts JT Jr. Par-athyroid hormone and parathyroid hormone-related peptide in the regula-tion of calcium homeostasis and bone development. In: DeGroot LJ, ed.Endocrinology. 4th ed. Philadelphia: W.B. Saunders (in press).2. Brown EM, Vassilev PM, Quinn S, Hebert SC. G-protein-coupled, ex-tracellular Ca2+-sensing receptor: a versatile regulator of diverse cellularfunctions. Vitam Horm 1999;55:1-71.3. Marchese A, George SR, Kolakowski LF, Lynch KR, ODowd BF. Nov-el GPCRs and their endogenous ligands: expanding the boundaries ofphysiology and pharmacology. Trends Pharmacol Sci 1999;20:370-5.[Erratum, Trends Pharmacol Sci 1999;20:447.]4. Demay MB, Kiernan MS, DeLuca HF, Kronenberg HM. Sequences inthe human parathyroid hormone gene that bind the 1,25-dihydroxyvitaminD3 receptor and mediate transcriptional repression in response to 1,25-dihydroxyvitamin D3. Proc Natl Acad Sci U S A 1992;89:8097-101.5. Moallem E, Kilav R, Silver J, Naveh-Many T. RNA-protein binding andpost-transcriptional regulation of parathyroid hormone gene expression bycalcium and phosphate. J Biol Chem 1998;273:5253-9.6.

    Chung U, Igarashi T, Nishishita T, et al. The interaction between Kuantigen and REF1 protein mediates negative gene regulation by extracel-lular calcium. J Biol Chem 1996;271:8593-8.7. Naveh-Many T, Rahamimov R, Livni N, Silver J. Parathyroid cell pro-liferation in normal and chronic renal failure rats: the effects of calcium,phosphate, and vitamin D. J Clin Invest 1995;96:1786-93.8. Li YC, Amling M, Pirro AE, et al. Normalization of mineral ion ho-meostasis by dietary means prevents hyperparathyroidism, rickets, and os-teomalacia, but not alopecia in vitamin D receptor-ablated mice. Endocri-nology 1998;139:4391-6.9. Malloy PJ, Pike JW, Feldman D. The vitamin D receptor and the syn-drome of hereditary 1,25-dihydroxyvitamin D-resistant rickets. Endocr Rev1999;20:156-88.10. Chorev M, Alexander JM, Rosenblatt M. Interactions of parathyroidhormone and parathyroid homonerelated protein with their receptors. In:Bilezikian JP, Levine MA, Marcus R , eds. The parathyroids: basic and clin-ical concepts. 2nd ed. San Diego, Calif.: Academic Press (in press).11. Juppner H. Receptors for parathyroid hormone and parathyroid hor-mone-related peptide: exploration of their biological importance. Bone1999;25:87-90.12. Usdin TB, Hoare SRJ, Wang T, Mezey E, Kowalak JA. TIP39: a newneuropeptide and PTH2-receptor agonist from hypothalamus. Nat Neuro-sci 1999;2:941-3.13. Strewler GJ. The physiology of parathyroid hormonerelated protein.N Engl J Med 2000;342:177-85.14. Kao PC, van Heerden JA, Grant CS, K lee GG, Khosla S. Clinical per-formance of parathyroid hormone immunometric assays. Mayo Clin Proc1992;67:637-45.15. Michelangeli VP, Heyma P, Colman PG, Ebeling PR. Evaluation of anew, rapid and automated immunochemiluminometric assay for the meas-urement of serum intact parathyroid hormone. Ann Clin Biochem 1997;34:97-103.16. Lepage R, Roy L, Brossard JH, et al. A non-(1-84) circulating para-thyroid hormone (PTH) fragment interferes significantly with intact PTHcommercial assay measurements in uremic samples. Clin Chem 1998;44:805-9.17. John MR, Goodman WG, Gao P, Cantor TL, Salusky IB, Juppner H.

    A novel immunoradiometric assay detects full-length human PTH but notamino-terminally truncated fragments: implications for PTH measure-ments in renal failure. J Clin Endocrinol Metab 1999;84:4287-90.18. Jaskowiak N, Norton JA, Alexander HR, et al. A prospective trial eval-uating a standard approach to reoperation for missed parathyroid adenoma.

    Ann Surg 1996;224:308-20.19. Irvin GL III, Molinari AS, Figueroa C, Carneiro DM. Improved suc-cess rate in reoperative parathyroidectomy with intraoperative PTH assay.

    Ann Surg 1999;229:874-8.20. Arnold A. Molecular basis of primary hyperparathyroidism. In: Bile-zikian JP, Levine MA, Marcus R, eds. The parathyroids: basic and clinicalconcepts. 2nd ed. San Diego, Calif.: Academic Press (in press).21. Arnold A, Brown MF, Urena P, Gaz RD, Sarfati E, Drueke TB. Mono-clonality of parathyroid tumors in chronic renal failure and in primary par-athyroid hyperplasia. J Clin Invest 1995;95:2047-53.22. Agarwal SK, Guru SC, Heppner C, et al. Menin interacts with the

    AP1 transcription factor JunD and represses JunD-activated transcription.Cell 1999;96:143-52.23. Malkin D. Li-Fraumeni syndrome. In: Scriver CR , Beaudet A, Sly WS,

    Valle D, Vogelstein B, eds. The metabolic and molecular bases of inheriteddisease. 8th ed. Vol. X. New York: McGraw-Hill (in press).24. Pestell RG, Albanese C, Reutens AT, Segall JE, Lee RJ, Arnold A. Thecyclins and cyclin-dependent kinase inhibitors in hormonal regulation ofproliferation and differentiation. Endocr Rev 1999;20:501-34.

    25. Masciullo V, Khalili K , Giordano A. The Rb family of cell cycle regu-latory factors: clinical implications. Int J Oncol 2000;17:897-902.26. Spiegel AM. Mutations in G proteins and G protein-coupled receptorsin endocrine disease. J Clin Endocrinol Metab 1996;81:2434-42.27. Hoff AO, Cote GJ, Gagel RF. Multiple endocrine neoplasias. AnnuRev Physiol 2000;62:377-411.28. Tahara H, Smith AP, Gaz RD, Cryns VL, Arnold A. Genomic local-ization of novel candidate tumor suppressor gene loci in human parathy-roid adenomas. Cancer Res 1996;56:599-605.29. Farnebo F, Kytola S, Teh BT, et al. Alternative genetic pathways in par-athyroid tumorigenesis. J Clin Endocrinol Metab 1999;84:3775-80.30. Kinzler KW, Vogelstein B. Lessons from hereditary colorectal cancer.Cell 1996;87:159-70.31. Knudson AG. Hereditary cancer: two hits revisited. J Cancer Res ClinOncol 1996;122:135-40.32. Cryns VL, Thor A, Xu H-J, et al. Loss of the retinoblastoma tumor-suppressor gene in parathyroid carcinoma. N Engl J Med 1994;330:757-61.33. Cryns VL, Rubio MP, Thor AD, Louis DN, Arnold A. p53 Abnor-

    malities in human parathyroid carcinoma. J Clin Endocrinol Metab 1994;78:1320-4.34. Agarwal SK, Schrock E, Kester MB, et al. Comparative genome hy-bridization analysis of human parathyroid tumors. Cancer Genet Cytogen-et 1998;106:30-6.35. Heppner C, Kester MB, Agarwal SK, et al. Somatic mutation of theMEN1gene in parathyroid tumours. Nat Genet 1997;16:375-8.36. Hosokawa Y, Pollak MR, Brown EM, Arnold A. Mutational analysisof the extracellular Ca(2+)-sensing receptor gene in human parathyroidtumors. J Clin Endocrinol Metab 1995;80:3107-10.37. Brown SB, Brierley TT, Palanisamy N, et al. Vitamin D receptor as acandidate tumor-suppressor gene in severe hyperparathyroidism of uremia.J Clin Endocrinol Metab 2000;85:868-72.38. Carling T, Szabo E, Bai M, et al. Familial hypercalcemia and hypercal-ciuria caused by a novel mutation in the cytoplasmic tail of the calcium re-ceptor. J Clin Endocrinol Metab 2000;85:2042-7.39. Motokura T, Bloom T, Kim HG, et al. A novel cyclin encoded by abcl1-linked candidate oncogene. Nature 1991;350:512-5.40. Hsi ED, Zukerberg LF, Yang WI, Arnold A. Cyclin D1/PRAD1 ex-pression in parathyroid adenomas: an immunohistochemical study. J ClinEndocrinol Metab 1996;81:1736-9.41. Brown EM, Pollak M, Hebert SC. The extracellular calcium-sensingreceptor: its role in health and disease. Annu Rev Med 1998;49:15-29.42. Wermers RA, Khosla S, Atkinson EJ, Hodgson SF, OFallon WM,Melton LJ III. The rise and fall of primary hyperparathyroidism: a popu-lation-based study in Rochester, Minnesota, 1965-1992. Ann Intern Med1997;126:433-40.43. Lundgren E, Rastad J, Thurfjell E, Akerstrom G, Ljunghall S. Popu-lation-based screening for primary hyperparathyroidism with serum calci-um and parathyroid hormone values in menopausal women. Surgery 1997;121:287-94.44. Schneider AB, Gierlowski TC, Shore-Freedman E, Stovall M, Ron E,Lubin J. Dose-response relationships for radiation-induced hyperparathy-roidism. J Clin Endocrinol Metab 1995;80:254-7.45. Bendz H, Sjodin I, Toss G, Berglund K. Hyperparathyroidism andlong-term lithium therapy a cross-sectional study and the effect of lith-ium withdrawal. J Intern Med 1996;240:357-65.46. Brown EM. Lithium induces abnormal calcium-regulated PTH releasein dispersed bovine parathyroid cells. J Clin Endocrinol Metab 1981;52:1046-8.47. Teh BT, Farnebo F, Twigg S, et al. Familial isolated hyperparathyroid-ism maps to the hyperparathyroidism-jaw tumor locus in 1q21-q32 in asubset of families. J Clin Endocrinol Metab 1998;83:2114-20.48. Kassem M, Kruse TA, Wong FK, Larsson C, Teh BT. Familial isolatedhyperparathyroidism as a variant of multiple endocrine neoplasia type 1 ina large Danish pedigree. J Clin Endocrinol Metab 2000;85:165-7.49. Marx SJ, Spiegel AM, Skarulis MC, Doppman JL, Collins FS, LiottaLA. Multiple endocrine neoplasia type 1: clinical and genetic topics. AnnIntern Med 1998;129:484-94.50. Marx SJ. Contrasting paradigms for hereditary hyperfunction of endo-crine cells. J Clin Endocrinol Metab 1999;84:3001-9.51. Schuffenecker I, Virally-Monod M, Brohet R, et al. Risk and pene-trance of primary hyperparathyroidism in multiple endocrine neoplasia

  • 8/12/2019 HIPER_HIPOPARATIR_NEJM

    12/13

    1874 December 21, 2000

    The New England Journal of Medicine

    type 2A families with mutations at codon 634 of the RETproto-oncogene.J Clin Endocrinol Metab 1998;83:487-91.52. Szabo J, Heath B, Hill VM, et al. Hereditary hyperparathyroidism-jawtumor syndrome: the endocrine tumor gene HRPT2 maps to chromosome1q21-q31. Am J Hum Genet 1995;56:944-50.53. Williamson C, Cavaco BM, Jauch A, et al. Mapping the gene causinghereditary primary hyperparathyroidism in a Portuguese kindred to chro-mosome 1q22-q31. J Bone Miner Res 1999;14:230-9. [Erratum, J Bone

    Miner Res 1999;14:1472.]54. Bilezikian JP, Silverberg SJ, Gartenberg F, et al. Clinical presentationof primary hyperparathyroidism. In: Bilezikian JP, Levine MA, Marcus R ,eds. The parathyroids: basic and clinical concepts. 2nd ed. San Diego, Cal-if.: Academic Press (in press).55. Silverberg SJ, Shane E, Jacobs TP, Siris E, Bilezikian JP. A 10-year pro-spective study of primary hyperparathyroidism with or without parathyroidsurgery. N Engl J Med 1999;341:1249-55. [Erratum, N Engl J Med 2000;342:144.]56. Chan AK, Duh QY, Katz MH, Siperstein AE, Clark OH. Clinicalmanifestations of primary hyperparathyroidism before and after parathy-roidectomy: a case-control study. Ann Surg 1995;222:402-14.57. Burney RE, Jones KR, Christy B, Thompson NW. Health status im-provement after surgical correction of primary hyperparathyroidism in pa-tients with high and low preoperative calcium levels. Surgery 1999;125:608-14.58. Steffenelli T, Abela C, Frank H, et al. Cardiac abnormalities in patients

    with primary hyperparathyroidism: implications for follow-up. J Clin En-docrinol Metab 1997;82:106-12.59.

    Wermers RA, Khosla S, Atkinson EJ, et al. Survival after diagnosis ofhyperparathyroidism: a population-based study. Am J Med 1998;104:115-22.60. Ishizuya T, Yokose S, Hori M, et al. Parathyroid hormone exerts dis-parate effects on osteoblastic differentiation depending on exposure timein rat osteoblastic cells. J Clin Invest 1997;99:2961-70.61. Schiller PC, DIppolito G, Roos BA, Howard GA. Anabolic or cata-bolic responses of MC3T3-E1 osteoblastic cells to parathyroid hormonedepend on time and duration of treatment. J Bone Miner Res 1999;14:1504-12.62. Dempster DW, Parisien M, Silverberg SJ, et al. On the mechanism ofcancellous bone preservation in postmenopausal women with mild primaryhyperparathyroidism. J Clin Endocrinol Metab 1999;84:1562-6.63. Abdelhadi M, Nordenstrom J. Bone mineral recovery after parathy-roidectomy in patients with primary and renal hyperparathyroidism. J ClinEndocrinol Metab 1998;83:3845-51.64. Silverberg SJ, Locker FG, Bilezikian JP. Vertebral osteopenia: a newindication for surgery in primary hyperparathyroidism. J Clin EndocrinolMetab 1996;81:4007-12.65. Khosla S, Melton LJ III, Wermers RA, Crowson CS, OFallon WM,Riggs BL. Primary hyperparathyroidism and the risk of fracture: a popula-tion-based study. J Bone Miner Res 1999;14:1700-7.66. Christiansen P, Steiniche T, Brixen K, et al. Primary hyperparathyroid-ism: effect of parathyroidectomy on regional bone mineral density in Dan-ish patients: a three-year follow-up study. Bone 1999;25:589-95.67. NIH conference: diagnosis and management of asymptomatic primaryhyperparathyroidism: Consensus Development Conference statement. AnnIntern Med 1991;114:593-7.68. Chen H, Parkerson S, Udelsman R. Parathyroidectomy in the elderly:do the benefits outweigh the risks? World J Surg 1998;22:531-6.69. Pattou F, Torres G, Mondragon-Sanchez A, et al. Correlation of par-athyroid scanning and anatomy in 261 unselected patients with sporadicprimary hyperparathyroidism. Surgery 1999;126:1123-31.70. Wei JP, Burke GJ. Cost utility of routine imaging with Tc-99m-sesta-mibi in primary hyperparathyroidism before initial surgery. Am Surg 1997;63:1097-100.71. Shawker TH, Avila N, Premkumar A, Bradford MH, Doppman JL.Ultrasound evaluation of primary hyperparathyroidism. Ultrasound Q2000;16:73-87.72. Norman J, Chheda H, Farrell C. Minimally invasive parathyroidectomyfor primary hyperparathyroidism: decreasing operative time and potentialcomplications while improving cosmetic results. Am Surg 1998;64:391-5.73. Ryan JA Jr, Lee F. Effectiveness and safety of 100 consecutive parathy-roidectomies. Am J Surg 1997;173:441-4.74. Low RA, Katz AD. Parathyroidectomy via bilateral cervical explora-tion: a retrospective review of 866 cases. Head Neck 1998;20:583-7.75. Stock JL, Marcus R. Medical management of primary hyperparathy-roidism. In: Bilezikian JP, Levine MA, Marcus R, eds. The parathyroids:basic and clinical concepts. 2nd ed. San Diego, Calif.: Academic Press (inpress).76. Brown DL, Robbins R. Developments in the therapeutic applicationsof bisphosphonates. J Clin Pharmacol 1999;39:651-60.77. Grey AB, Stapleton JP, Evans MC, Tatnell MA, Reid IR. Effect of hor-

    mone replacement therapy on bone mineral density in postmenopausalwomen with mild primary hyperparathyroidism: a randomized, controlledtrial. Ann Intern Med 1996;125:360-8.78. Silverberg SJ, Bone HG III, Marriott TB, et al. Short-term inhibitionof parathyroid hormone secretion by a calcium-receptor agonist in patients

    with primary hyperparathyroidism. N Engl J Med 1997;337:1506-10.79. Antonsen JE, Sherrard DJ, Andress DL. A calcimimetic agent acutelysuppresses parathyroid hormone levels in patients with chronic renal fail-

    ure: rapid communication. Kidney Int 1998;53:223-7.80. Parfitt AM. The hyperparathyroidism of chronic renal failure: a disor-der of growth. Kidney Int 1997;52:3-9.81. Malberti F, Farina M, Imbasciati E. The PTH-calcium curve and theset point of calcium in primary and secondary hyperparathyroidism. Neph-rol Dial Transplant 1999;14:2398-406.82. Shan L, Nakamura Y, Murakami M, et al. Clonal emergence in uremicparathyroid hyperplasia is not related to MEN1 gene abnormality. Jpn JCancer Res 1999;90:965-9.83. Imanishi Y, Tahara H, Salusky I, et al. MEN1gene mutations in re-fractory hyperparathyroidism of uremia. J Bone Miner Res 1999;14:Suppl1:S446. abstract.84. Horandner HH, Neyer U, Gruber U, et al. Pathomorphology of par-athyroid autografts. Nieren Hochdruckkrankheiten 1997;26:319-27.85. Solal M-EC, Sebert J-L, Boudailliez B, et al. Comparison of intact,midregion, and carboxy terminal assays of parathyroid hormone for the di-agnosis of bone disease in hemodialyzed patients. J Clin Endocrinol Metab1991;73:516-24.86. Sherrard DJ, Hercz G, Pei Y, et al. The spectrum of bone disease in

    end-stage renal failure an evolving disorder. Kidney Int 1993;43:436-42.87. Mucsi I, Hercz G. Relative hypoparathyroidism and adynamic bonedisease. Am J Med Sci 1999;317:405-9.88. Wang M, Hercz G, Sherrard DJ, Maloney NA, Segre GV, Pei Y. Re-lationship between intact 1-84 parathyroid hormone and bone histomor-phometric parameters in dialysis patients without aluminum toxicity. Am JKidney Dis 1995;26:836-44.89. Hampl H, Steinmuller T, Frohling P, et al. Long-term results of totalparathyroidectomy without autotransplantation in patients with and with-out renal failure. Miner Electrolyte Metab 1999;25:161-70.90. Hasnain M, Hauncher C, Pegoraro AA, Arruda JAL, Dunea G. Sup-pression of hyperparathyroidism by calcitriol therapy. ASAIO J 1999;45:424-7.91. Bacchini G, Fabrizi F, Pontoriero G, Marcelli D, Filippo SD, LocatelliF. Pulse oral versus intravenous calcitriol therapy in chronic hemodialysispatients: a prospective and randomized study. Nephron 1997;77:267-72.[Erratum, Nephron 1998;79:509.]92. Loghman-Adham M. Phosphate binders for control of phosphate re-tention in chronic renal failure. Pediatr Nephrol 1999;13:701-8.93. Indridason OS, Quarles LD. Comparison of treatments for mild sec-ondary hyperparathyroidism in hemodialysis patients. Kidney Int 2000;57:282-92.94. Tominaga Y. Surgical management of secondary hyperparathyroidismin uremia. Am J Med Sci 1999;317:390-7.95. Torres A, Rodriguez AP, Concepcion MT, et al. Parathyroid functionin long-term renal transplant patients: importance of pre-transplant PTHconcentrations. Nephrol Dial Transplant 1998;13:Suppl 3:94-7.96. Kerby JD, Rue LW, Blair H, Hudson S, Sellers MT, Diethelm AG. Op-erative treatment of tertiary hyperparathyroidism: a single-center experi-ence. Ann Surg 1998;227:878-86.97. Novelli A, Sabani M, Caiola A, et al. Diagnosis of DiGeorge and Wil-liams syndromes using FISH analysis of peripheral blood smears. Mol CellProbes 1999;13:303-7.98. Markert ML, Boeck A, Hale LP, et al. Transplantation of thymus tissuein complete DiGeorge syndrome. N Engl J Med 1999;341:1180-9.99. Ding C, Buckingham B, Levine MA. Neonatal hypoparathyroidism at-tributable to homozygous partial deletion of the human glial cell missinggene-B. In: Program and abstracts of the 82nd Annual Meeting of the En-docrine Society, Toronto, June 2124, 2000. Bethesda, Md.: EndocrineSociety Press, 2000:409. abstract.100. Ahonen P, Myllrniemi S, Sipil I, Perheentupa J. Clinical variationof autoimmune polyendocrinopathycandidiasisectodermal dystrophy(APECED) in a series of 68 patients. N Engl J Med 1990;322:1829-36.101. Bjorses P, Halonen M, Palvimo JJ, et al. Mutations in the AIREgene:effects on subcellular location and transactivation function of the autoim-mune polyendocrinopathy-candidiasis-ectodermal dystrophy protein. Am JHum Genet 2000;66:378-92.102. Sunthornthepvarakul T, Churesigaew S, Ngowngarmratana S. A novelmutation of the signal peptide of the preproparathyroid hormone gene as-sociated with autosomal recessive familial isolated hypoparathyroidism.J Clin Endocrinol Metab 1999;84:3792-6.103. Pearce SHS, Williamson C, Kifor O, et al. A familial syndrome of hy-

  • 8/12/2019 HIPER_HIPOPARATIR_NEJM

    13/13

    MEDICAL PROGRESS

    Vol ume 343 Nu mb er 25 1875

    pocalcemia with hypercalciuria due to mutations in the calcium-sensing re-ceptor. N Engl J Med 1996;335:1115-22.104. Winer KK, Yanovski JA, Cutler GB Jr. Synthetic human parathyroidhormone 1-34 vs calcitriol and calcium in the treatment of hypoparathy-roidism. JAMA 1996;276:631-6.105. Schipani E, Langman CB, Parfitt AM, et al. Constitutively activatedreceptors for parathyroid hormone and parathyroid hormonerelated pep-tide in Jansens metaphyseal chondrodysplasia. N Engl J Med 1996;335:

    708-14.106. Zhang P, Jobert AS, Couvineau A, Silve C. A homozygous inactivat-ing mutation in the parathyroid hormone/parathyroid hormone-related

    peptide receptor causing Blomstrand chondrodysplasia. J Clin EndocrinolMetab 1998;83:3365-8.107. Davies SJ, Hughes HE. Imprinting in Albrights hereditary osteodys-trophy. J Med Genet 1993;30:101-3.108. Yu SH, Yu DW, Lee E, et al. Variable and tissue-specific hormoneresistance in heterotrimeric Gs protein alpha subunit (Gsalpha) knockoutmice is due to tissue-specific imprinting of the Gsalpha gene. Proc Natl

    Acad Sci U S A 1998;95:8715-20.

    109. Liu J, Litman D, Rosenberg MJ, Yu S, Biesecker LG, Weinstein LS.A GNAS1imprinting defect in pseudohypoparathyroidism type IB. J ClinInvest 2000;106:1167-74.