Skip to main content
Erschienen in: Orphanet Journal of Rare Diseases 1/2008

Open Access 01.12.2008 | Review

McCune-Albright syndrome

verfasst von: Claudia E Dumitrescu, Michael T Collins

Erschienen in: Orphanet Journal of Rare Diseases | Ausgabe 1/2008

Abstract

McCune-Albright syndrome (MAS) is classically defined by the clinical triad of fibrous dysplasia of bone (FD), café-au-lait skin spots, and precocious puberty (PP). It is a rare disease with estimated prevalence between 1/100,000 and 1/1,000,000. FD can involve a single or multiple skeletal sites and presents with a limp and/or pain, and, occasionally, a pathologic fracture. Scoliosis is common and may be progressive. In addition to PP (vaginal bleeding or spotting and development of breast tissue in girls, testicular and penile enlargement and precocious sexual behavior in boys), other hyperfunctioning endocrinopathies may be involved including hyperthyroidism, growth hormone excess, Cushing syndrome, and renal phosphate wasting. Café-au-lait spots usually appear in the neonatal period, but it is most often PP or FD that brings the child to medical attention. Renal involvement is seen in approximately 50% of the patients with MAS. The disease results from somatic mutations of the GNAS gene, specifically mutations in the cAMP regulating protein, Gs alpha. The extent of the disease is determined by the proliferation, migration and survival of the cell in which the mutation spontaneously occurs during embryonic development. Diagnosis of MAS is usually established on clinical grounds. Plain radiographs are often sufficient to make the diagnosis of FD and biopsy of FD lesions can confirm the diagnosis. The evaluation of patients with MAS should be guided by knowledge of the spectrum of tissues that may be involved, with specific testing for each. Genetic testing is possible, but is not routinely available. Genetic counseling, however, should be offered. Differential diagnoses include neurofibromatosis, osteofibrous dysplasia, non-ossifying fibromas, idiopathic central precocious puberty, and ovarian neoplasm. Treatment is dictated by the tissues affected, and the extent to which they are affected. Generally, some form of surgical intervention is recommended. Bisphosphonates are frequently used in the treatment of FD. Strengthening exercises are recommended to help maintaining the musculature around the FD bone and minimize the risk for fracture. Treatment of all endocrinopathies is required. Malignancies associated with MAS are distinctly rare occurrences. Malignant transformation of FD lesions occurs in probably less than 1% of the cases of MAS.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1750-1172-3-12) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.
Abkürzungen
FD
fibrous dysplasia of bone
PP
precocious puberty
GNAS
G protein binding adenylyl cyclase stimulatory
cAMP
cyclic adenosine monophosphate
Gs alpha
G protein α-subunit
MAS
McCune-Albright syndrome
GH
growth hormone
NF
neurofibromatosis
NOF
Non-ossifying fibromas
CF
craniofacial
TSH
thyroid stimulating hormone
FT4
free thyroxine
T3
triiodothyronine
T4
thyroxine
FGF-23
fibroblast growth factor-23
TRP
tubular reabsorption of phosphate
TmP/GFR
maximal tubular reabsorption of phosphate per glomerular filtration rate
OGTT
oral glucose tolerance test
MRI
magnetic resonance imaging
CT
computed tomography
IGF-1
insulin-like growth factor-1
PTH
parathyroid hormone
MSH
melanocyte stimulating hormone
LH
luteinizing hormone
GHRH
growth hormone stimulating hormone
ACTH
adrenocrotical stimulating hormone
E2
estradiol
T
testosterone
99 Tc-MDP
Technetium 99 methylene diphosphonate

Definition

Originally, the McCune-Albright syndrome (MAS) was defined by the triad of polyostotic fibrous dysplasia of bone (FD), café-au-lait skin pigmentation, and precocious puberty (PP) [1, 2]. It was later recognized that other endocrinopathies, including hyperthyroidism (reviewed in [3]), growth hormone (GH) excess [4, 5], renal phosphate wasting with or without rickets/osteomalacia [6] and Cushing syndrome could be found in association with the original triad [79]. Rarely, other organ systems may be involved (liver, cardiac, parathyroid, pancreas) [10].
While MAS is rare, FD is not. FD can involve a single skeletal site (monostotic FD, MFD), or multiple sites (polyostotic FD, PFD) [1114]. Very rarely PP can be found in association with café-au-lait skin pigmentation in the absence of FD (about 1% of the cases), but in general, FD seems to be the most common component of MAS. Therefore, a more clinically relevant definition of MAS, broader than the original triad of FD + PP + café-au-lait is: MAS = FD + at least one of the typical hyperfunctioning endocrinopathies and/or café-au-lait spots, with almost any combination possible [13, 15].

Epidemiology

MAS is a rare disease and reliable data of prevalence are not available (the estimated prevalence ranges between 1/100,000 and 1/1,000,000). In contrast, the skeletal aspect of the disease, FD, especially monostotic disease, is not rare [16]. FD has been reported to account for up to 7% of all benign bone tumors.

Clinical description

Typically, the signs and symptoms of either PP or FD usually account for the initial presentation. In girls with PP, it is usually vaginal bleeding or spotting, accompanied by development of breast tissue, usually without the development of pubic hair. In boys, it can be bilateral (or unilateral) testicular enlargement with penile enlargement, scrotal rugae, body odor, pubic and axillary hair, and precocious sexual behavior. In retrospect, café-au-lait spots (Fig. 1), which are usually present at birth or shortly thereafter, are the most common but unappreciated "presenting" sign.
Fibrous dysplasia in the appendicular skeleton usually presents with a limp and/or pain (sometimes reported by children as being "tired"), but occasionally a pathologic fracture may be the presenting sign. Radiographs will demonstrate typical expansile lesions with endosteal scalloping and thinning of the cortex with the matrix of the intramedullary tissue demonstrating a "ground glass" appearance (Fig. 2A &2B). FD in the craniofacial bones usually presents as a painless "lump" or facial asymmetry. Representative radiographic findings and the histological appearance of FD are shown in Figures 2 and 3. The areas most commonly involved are the proximal femora and skull base. The sites of FD involvement are established early; 90% of the total body skeletal disease burden is usually established by age 15 [17]. Hart et al. found that lesions in the craniofacial region were established earliest, with 90% of the lesions present by 3.4 years of age. In the extremities, 90% were present by 13.7 yr, and in the axial skeleton, 90% were present by 15.5 yr. The appearance of new lesions later in life is a very uncommon occurrence in FD. The incidence of fractures is greatest in childhood, between the age of 6 and 10 yr, but due to the intrinsic abnormalities in FD bone, some fractures continue to occur into adulthood (Fig. 4)[18].
Other features of the presentation related to the specific aspect of the disease are outlined in Table 1.
Table 1
Clinical presentation
Clinical feature
Presentation/Feature
Café-au-lait spots
appearance: "coast-of-Maine", location: nape of neck, base of spine; usually respect midline; trunk & face more commonly involved than limbs (Figure 1)
Fibrous dysplasia (FD)
phosphatase, medullary cavity of the bone. [16, 18]
Craniofacial FD
nerve decompression is contraindicated [40, 41]
Axial FD
usually asymptomatic, scoliosis common and possibly progressive [56] rib pain develops late
Appendicular FD
often painful in adults, but pain is quite variable [17, 18]
Gonadal – Female
Vaginal bleeding, breast development with little or no pubic hair, secondary central PP often develops, GnRH test [66]
Gonadal – Male
common, detectable by ultrasound only, cancer rare, but possible [67]
Thyroid abnormalities
ultrasound and biopsy large solid or changing lesions [3], cancer can occur [24]
Phosphaturia
worsens pain [6]; etiology is FGF23 of FD bone origin [43]
Growth hormone (GH) excess
suggests GH excess, worsens craniofacial FD [5, 41]
Cushing syndrome
quite ill, spontaneous resolution possible [9]
Additional clinical features
alopecia [70], myxomas and others cutaneous abnormalities [71]

Malignancies in MAS

While malignancies associated with MAS are distinctly rare occurrences, they warrant mentioning due to their importance. Malignant transformation of FD lesions is probably the most common and best described malignancy that occurs in association with MAS [12, 16, 19]. This occurs in probably less than 1% of the cases of FD/MAS. High dose external beam radiation is a risk factor for sarcomatous transformation [19]. There may be a greater tendency for malignant transformation to occur in patients who have concomitant GH excess [20]. While some have suggested that sarcomatous transformation of skeletal lesions may occur more commonly in patients with Mazabraud's syndrome (benign intramuscular myxomas in association with long standing FD) [21], this may represent selection bias.
In addition, the risk of breast cancer may be elevated in patients with MAS [22, 23]. Besides the published reports, in the series of approximately 120 patients seen at the National Institutes of Health (NIH), the prevalence of breast cancer was approximately 2.5% (n = 3). In this group of three patients, there also seems to be an effect of GH excess, in that all patients who had breast cancer also had GH excess (unpublished data).
Thyroid cancer [24] and testicular cancer (unpublished data) are also rare occurrences.

Etiology

The observation that the G protein/cAMP/adenylate cyclase signaling pathway was central to all of the tissues involved in MAS eventually led to the discovery that mutations in the regulatory Gsα protein (encoded by the GNAS gene) were the underlying molecular etiology of MAS [25, 26] (Fig. 5). In all published cases of MAS, PFD, and even MFD, activating mutations of Gsα at the R201 position have been identified [27]. More recently, mutations at the Q227 position have been found in association with FD [28].
The lack of vertical transmission of the disease, along with the observation that skin and bone lesions tend to respect the midline and be on one or the other side of the body, has led to the unproven, but accepted, concept that the disease is the result of postzygotic mutations, and that patients are therefore somatic mosaics. The point in time in development at which the mutation occurs, the specific cell in which it occurs, and to where its progeny migrate, determines what tissues will be affected, and thus the phenotype. Therefore, in cases in which tissues of endodermal, mesodermal, and ectodermal origin are involved, it would appear that the mutation occurred at the inner cell mass stage (Fig. 6) [29].

Diagnosis, diagnostic criteria, diagnostic methods, differential diagnosis

Diagnosis of MAS is usually established on clinical grounds. Plain radiographs are often sufficient to make the diagnosis of FD (Fig. 2). Isotopic bone scans are the most sensitive tool for detecting the presence of FD lesions, and are often useful, especially at the initial evaluation, for determining the extent of the disease and predicting functional outcome (Fig. 4E,F, &4G) [17, 30]. FD has a typical appearance on radiographs described as "ground glass." In general, lesions in the long bones have a "lytic" appearance. The lesions usually arise in the medullary cavity and expand outward replacing normal bone, which results in thinning of the cortex (Fig. 2A &2B). It is usually the metaphysis and/or the diaphysis that are involved, with sparing of the epiphysis. It is possible for any bone to be involved, but the skull base and the proximal femur are the sites most commonly involved [16, 31, 32]. Due to the fact that these lesions are undermineralized [27], the bones are "soft" and prone to deformation, as exemplified by the classic "shepherd's crook" deformity of the proximal femur (Fig. 2A).
FD in the craniofacial bones tends to have a "sclerotic" appearance on plain radiographs. This is due to the relatively greater degree of mineralization of FD tissue in the craniofacial bones (Fig. 3) [33]. Computed tomography (CT) scanning is the best technique for imaging FD lesions in the skull, revealing a "ground glass" appearance. In children and young adults, the lesions appear homogeneous on CT, but in older patients the appearance is mixed, with the development of "cystic" lesions in some areas. The density of these areas is that of soft tissue, so while they may have a cystic appearance they are not true cysts. That said, it is possible for true cysts to develop in FD, both in the long bones, but more often in the craniofacial bones (Fig. 7). This has occurred in about 5% of the patients with FD in the NIH cohort (unpublished data). If needed, bone cysts may be diagnosed using magnetic resonance imaging (MRI). The cysts tend to have a more aggressive course. They can expand rapidly and produce symptoms which vary, depending on the location. One of the complications can be the fracture through a cyst. This usually requires surgical intervention.
Biopsy of FD lesions can confirm the diagnosis if doubt remains after review of the radiographs. One characteristic of FD bone is the absence of the lamellation pattern seen in normal bone under polarized light. This indicates that the matrix produced in the lesions is of the woven type. The histopathological description of FD is often described as a "Chinese writing" pattern, and with special preparation and stains used to detect mineralized and unmineralized tissue, extensive areas of unmineralized osteoid are evident (Fig. 3) [27]. For an extensive description of the histopathological changes that can be observed in FD, the reader is referred to Riminucci et al. and Corsi et al. [33, 34].

Genetic testing

Genetic testing is possible, but is not routinely available (see below). Because of the somatic mosaic nature of the disease, a negative result from readily available (but unaffected) tissue does not exclude the presence of the mutation. Testing on leukocyte DNA is possible [35], but it is unreliable. In most cases, genetic testing contributes little to the diagnosis and not at all to management. There is no known genotype/phenotype correlation, so knowledge of the specific mutation does not affect management. For this reason, when the diagnosis of MAS is suspected or established, it is important to be cognizant of the spectrum of tissues that possibly can be involved, and to screen for involvement. Screening, at a minimum, requires a medical history and physical examination, and usually involves specific imaging and biochemical testing.
The following are listed as potential sources for genetic testing: Center for Genetic Testing at Saint Francis [36], Genome Diagnostics [37]. In addition, the research laboratories of Professors Francis Glorieux, Shriners Hospital for Children (Montreal, Canada), Paolo Bianco (Rome, Italy) and Charles Sultan (Montpellier, France) can perform genetic testing on a research basis.

Differential diagnosis

MAS is most commonly confused with neurofibromatosis (NF), usually when a child presents with a large café-au-lait spot. The location and shape of the spots usually can help to distinguish between the MAS and NF. The spots in MAS have jagged borders (coast of Maine), whereas those in NF are smooth (coast of California). Although the spots can cross the midline, more often, they demonstrate a "respect" of the midline. Frequent locations are the nape of the neck and the crease at the apex of the buttocks (Fig. 1). In MAS, the skeletal disease (PFD) almost always involves one or both proximal femurs and/or the skull base, as well as other locations. Skeletal involvement in NF is uncommon and usually involves the diaphyses of the long bones, especially the tibiae, often leading to pseudoarthrosis [38, 39].
When precocious puberty is the presenting sign, the differential diagnosis includes idiopathic central precocious puberty, and an ovarian neoplasm. Suppressed gonadotropins exclude central PP. Other components of the MAS (skin pigmentation, skeletal changes on x-ray or bone scan, etc.) can help to assure the clinician that the ovarian cyst is not neoplastic.
Isolated skeletal lesions in the absence of skin or endocrine findings represent FD (MFD or PFD). Osteofibrous dysplasia (ossifying fibroma of long bones, so-called Campanacci's lesion) may be confused with FD. These lesions are almost exclusively found in the tibia and fibula, and are histologically distinct from FD [12]. Non-ossifying fibromas (NOF) may also share radiological and histological similarities with FD in the long bones. Lack of multiple skeletal foci and absence of extraskeletal findings may help to distinguish FD from NOF. FD in the jaw may share several histological features with cemento ossifying fibromas, which can be confused with FD. The cemento ossifying fibroma lesions tend to behave more aggressively than do FD lesions. Testing for the GNAS mutation, if tissues and assays are available, may be helpful in distinguishing cemento ossifying fibroma lesions from FD.

Specific diagnostic considerations and follow-up

A. Skeletal

Skeletal disease, especially involving the skull base, is very common. Vision and/or hearing loss are uncommon, and sarcomatous transformation is rare (<1%). FD in the appendicular skeleton tends to quiet with age, but the course of craniofacial disease is less clear [18, 40]. Nuclear medicine bone scans are useful for not only detecting the extent of the disease, but quantifying the skeletal disease burden of FD and predicting functional outcome [30].

1. Craniofacial FD

CT scan of the skull is the most useful test for diagnosis of craniofacial FD (Fig. 4). CT should be repeated periodically, annually then less frequently once stability has been demonstrated. Vision should be evaluated, ideally by a neuro-ophthalmologist initially, then periodically; annually or less frequently once stability has been demonstrated. Hearing evaluation is recommended at baseline to assess involvement and should also be repeated periodically. All endocrinopathies which adversely affect bone should be screened for and treated. GH excess in particular may adversely affect craniofacial FD [5, 40, 41]. 99Tc-methyl diphosphonate (99Tc-MDP) bone scan at baseline and periodically is recommended.

2. Axial and appendicular skeleton

Plain radiographs, demonstrating the classic ground glass appearance, are usually sufficient for the diagnosis of FD in the axial and appendicular skeleton (the axial skeleton is comprised of bones of the skull, hyoid bone, vertebra, sternum and ribs; the appendicular skeleton includes the hip, pelvic bone and the shoulder girdle (clavicle and scapula) (Fig. 2). However, x-rays are often unable to detect new, small microfractures. When new focal pain develops in an FD lesion and no fracture is evident on plain radiograph, CT and or MRI can be useful for detecting subtle fractures (Fig. 8). The classic lesion of the proximal femur in FD, the shepherd's crook deformity is common (Fig. 2).

B. Endocrine

1. Gonads

PP is more common in girls then boys. However, small testicular masses of Leydig and/or Sertoli cell hyperplasia are common in boys. These are usually never detected on physically examination. Therefore, ultrasound of the testes is recommended in all males with FD and MAS. It is probably safe to monitor testicular masses without surgical intervention as cancer is probably quite rare. The PP of MAS in girls is due to high levels of serum estradiol due to intermittent autonomous activation of ovarian tissue. Some of them will progress to central PP, secondary to activation of the hypothalamic-pituitary-gonadal (HPG) axis. Since central PP usually develops in children who have had PP for some years, children with PP should be evaluated for secondary central PP.

2. Thyroid

Hyperthyroidism is common (38%) [3]. Evidence of thyroid involvement without frank hyperthyroidism is even more common (63% in the NIH series, unpublished data). This is manifested as a relatively suppressed thyroid stimulating hormone (TSH) with elevated Triiodothyronine (T3+) and an abnormal thyroid gland on ultrasound. Some of these patients may go on to develop frank hyperthyroidism, therefore, follow-up with measurement of TSH, free thyroxine (FT4), T3, and thyroxine (T4), and periodic ultrasound is recommended to detect those patients that progress to frank hyperthyroidism. While the development of thyroid cancer in patients with MAS is rare [24], patients should be monitored for this possibility. This is accomplished by annual ultrasound of the thyroid. The ultrasonographic appearance of the thyroid in MAS is complicated [42]. There are usually admixed areas of relatively normal appearing thyroid with adjacent cystic areas. Suspicion for cancer should be raised if a large or expanding solid lesion is noted. This should prompt fine needle aspiration with cytopathological evaluation.

3. Renal phosphate wasting

Renal phosphate wasting, as part of a proximal tubulopathy, with or without hypophosphatemia, and/or rickets/osteomalacia is common [6]. The etiology is likely due to elaboration of the phosphaturic factor, fibroblast growth factor-23 (FGF23), by FD tissue [43]. Measurement of serum phosphate and calculation of renal phosphate handling by either the tubular reabsorption of phosphate (TRP) or maximal tubular reabsorption of phosphate per glomerular filtration rate (TmP/GFR) are recommended. While it is debatable whether or not hypophosphatemia, per se, should be treated, or if treatment should be reserved for only patients with rickets or biopsy-proven rickets [44, 45], it is our feeling that frank hypophosphatemia should be treated (see Additional file 1).

4. Pituitary

GH and prolactin excess are common (21%), and the signs and symptoms can be very subtle. Since GH excess can worsen craniofacial bone disease [5, 40, 41], it is important to make the diagnosis and treat. All patients should have an oral glucose tolerance test (OGTT) to look for non-suppressible GH at least once. Most GH secreting tumors are co-secretors of GH and prolactin, so prolactin levels should also be assessed, as hyperprolactinemia can independently have an adverse affect on gonadal function.

5. Parathyroid

Primary hyperparathyroidism in MAS is rare and is probably not a part of the syndrome [46]. Secondary hyperparathyroidism, usually due to vitamin D deficiency is common in the general population as well as in FD/MAS [4750]. Hyperparathyroidism can worsen FD and should be treated [34]. Total or ionized serum calcium and parathyroid hormone (PTH) need to be assessed periodically.

6. Adrenal

Cushing syndrome can occur in the neonatal period, but has not been reported past the first year. Some cases of neonatal Cushing resolve spontaneously [9]. Cushing syndrome must be considered and screened for in very young patients. The physical examination is usually suggestive and shows moon facies with plethora, hirsutism, and lack of linear growth (Fig. 9). If suspected, laboratory evaluation with some combination of a 24-hour urine for urinary free cortisol (if possible), midnight, or salivary cortisol, and/or dexamethasone suppression test are indicated. Adrenal reserve should be checked in patients in whom Cushing resolved spontaneously (personal comment, in the NIH series 2 out of 9 patients with Cushing had spontaneously cure and inadequate adrenal reserve later in life).

Genetic counseling

The most important aspect of counseling families is to assure the patients or families that there is no vertical transmission of the disease, nor are there any known environmental associations or predilection for ethnic groups. Therefore, parents need not feel "responsible," and patients can be assured they will not transmit the disease to their offspring.

Antenatal diagnosis

Not relevant.

Management, treatment, and prognosis

A. Skeletal

1. Craniofacial FD

In the vast majority of cases of craniofacial FD, surgery is not needed, and observation is the correct approach. Observation involves annual vision and hearing testing, and imaging. Vision should be checked by a neuro-ophthalmologist. CT of skull and/or mandible is the imaging test of choice for the cranial structures in general, but MRI will give better resolution of the optic nerves. Cranial nerves are often surrounded by FD bone, but remain unaffected for years to decades [41]. Indications for surgery include documented progressive visual disturbance, severe pain, or severe disfigurement. Surgery should be avoided in the absence of visual or hearing impairment. However, when surgery is indicated, it is very important to find a craniofacial and neurosurgical team experienced in treating craniofacial FD. It is possible the craniofacial disease may continue to progress slowly into adulthood, but this subject has not been well studied in a prospective manner [40].
There is little evidence that bisphosphonates are effective in craniofacial FD, even for pain, but in the case of pain that is difficult to manage, bisphosphonate treatment should be considered (Additional file 2).

2. Axial and appendicular skeleton

The surgical management of FD has evolved over the years. While curettage and bone grafting were once considered the treatment of choice, experts in the care of FD no longer feel this is effective [31, 51]. Intramedullary devices are preferable, in general [31, 51]. Bracing may be helpful, but has not been well-studied. Shepherd's crook deformity can develop, sometimes very rapid, and the management in growing children is very challenging. Generally, some form of surgical intervention is recommended. Screening for and treatment of all endocrinopathies which adversely affect the skeleton should be performed [34]. This includes PP, hyperthyroidism, Cushing's syndrome, GH excess, and secondary hyperparathyroidism.
Bisphosphonates are frequently used in the treatment of FD (reviewed in [45, 52]. The original thinking in the use of bisphosphonates in FD was that they would prevent the progression of disease [53]. However, more recent work concludes that bisphosphonates have no effect on the natural history of the disease [54, 55]. It is clear, however, that bisphosphonates are effective in relieving bone pain associated with FD [32, 45, 52]. Many regimens have been suggested. The long term safety of bisphosphonates, especially in children, and the effects of relatively high doses of bisphosphonates on normal bone not affected by FD, are unknown. Therefore, to minimize exposure, our approach has been to use the minimum dose with the longest interval between doses possible needed to achieve and maintain pain relief (Additional file 2).
Maintaining the musculature around the FD bone is important for protecting the bone. Therefore, strengthening exercises and strength maintenance is important. Swimming and cycling are excellent modes of exercise for patients with FD that will promote strengthening and minimize the risk for fracture.
Scoliosis is common, and may be progressive [56]. Therefore, it is important to screen for the presence of scoliosis and monitor for any progression. Significant progression can occur within a short period of time.

B. Endocrine

1. Gonads

The management of PP in boys and girls is detailed in Additional file 3. The class of drugs with the longest history of safety and efficacy are the aromatase inhibitors [57, 58]. More recently, the estrogen antagonist/agonist, tamoxifen, has also shown promise [59]. Secondary central PP in children with MAS frequently develops after a period of PP. Its manifestation is often pubertal progression in a child that had previously been well-controlled on medical therapy. The treatment of secondary central PP in children with MAS is the same as in children with idiopathic PP, and involves the use of long-acting gonadotropin releasing hormone analogues.
Ultrasound is used to screen and follow boys and men with testicular masses. These are usually Leydig tumors, but testes can demonstrate a mixture of Leydig and Sertoli cell masses. Microlithiasis is also commonly seen. The development of malignancy in the testes of men with MAS appears to be very rare. Therefore, prudent management may be observation with annual testicular ultrasounds, and CT of the chest abdomen and pelvis if there is any suspicion for malignancy. Lesions should be followed annually with ultrasound. There are no reported cases of gynecological malignancy in females with MAS.

2. Thyroid

Antithyroidal medications, such as thionamides, are usually effective in controlling hyperthyroidism in MAS [60]. Spontaneous resolution of hyperthyroidism in MAS almost never occurs, so some form of definitive treatment, either in the form of surgery or radiation, is eventually indicated. Periodic ultrasounds of the thyroid to follow lesions should be performed annually. If a dominant or changing lesion is identified, fine needle aspiration is indicated to exclude cancer, given that thyroid cancer can be seen in the setting of MAS [24].

3. Renal

Renal involvement, in the form of renal phosphate wasting, is seen in approximately 50% of the patients with MAS [6]. However, only some of those patients will have a degree of phosphate wasting that results in hypophosphatemia (18% of the total population seen at NIH). It is debatable whether the hypophosphatemia warrants treatment with oral phosphorus supplementation if signs of rickets are absent [44]. However, we recommend that patients with frankly low serum phosphate should be treated. Treatment is the same as that for patients with X-linked hypophosphatemia and tumor induced osteomalacia: high dose of oral phosphate with high dose of calcitriol (Additional file 1).

4. Pituitary

GH excess is seen in approximately 20% of the patients with MAS [5]. GH excess has been shown to be associated with vision and hearing loss, and macrocephaly in patients with MAS [41]. For that reason, patients with GH excess and an elevated insulin-like growth factor-1 (IGF-1) should be treated. Treatment options include medications, surgery, and radiation. Effective medical treatment includes long-acting somatostatin analogues [4, 5] and the GH receptor antagonist, pegvisomant [6163]. There is greater long-term efficacy and safety with the somatostatin analogues, especially in children. Pegvisomant is significantly more expensive than the somatostatin analogues and may be associated with more side effects [61]. The goal of treatment in a growing child is to reduce the calculated serum IGF-1 Z-score to 0.
Surgery is an option, but is sometimes not possible due to the massive thickening of the skull bones. A trans-sphenoidal approach is the preferred method in pituitary surgery, but the skull base is virtually always involved with FD in patients with MAS and GH excess, so this approach is often impossible, or difficult, at best. A trans-frontal approach is a less desirable approach to the pituitary, in general, and in MAS it is also usually complicated by FD. Furthermore, in patients with MAS and GH excess the pituitary gland is almost always diffusely involved with areas of hyperplasia and adenoma (Edward Oldfield, personal communication). Therefore, the only hope for "cure" of the GH excess is a total hypophysectomy. This may be the best treatment in some cases, but physicians, surgeons, and patients should embark upon this option knowing that the patient will be rendered panhypopituitary at the end of the operation.
Radiotherapy is an option when surgery is not possible and medicinal treatment fails. However, there may be a higher risk of sarcomatous transformation [19, 62].
Since most of the GH secreting tumors co-secrete prolactin [5], dopamine agonists (cabergoline, etc.) are usually necessary, and are effective at normalizing the serum prolactin.

5. Parathyroid

While primary hyperparathyroidism has been reported to be part of the syndrome in the past [64], this may not be the case. When the mutations that cause MAS were sought in parathyroid tissue, they were not present [46]. Additionally, parathyroid hormone secretion and hyperplasia is likely not a cAMP, Gsα mediated phenomenon. Secondary hyperparathyroidism due to vitamin D deficiency is common and can worsen FD [34]. Therefore, it should be screened for and treated.

6. Adrenal

Cushing syndrome is one of the rarest, but most profound manifestations of MAS. In some cases, it can resolve spontaneously [9], but in most cases treatment is required, and in some cases, in spite of the best of care, it can be fatal. Adrenalectomy is probably the treatment of choice. If the child is too ill, adrenal blockade may provide stabilization of the disease and buy time until a point in time when surgery can be performed. Liver function abnormalities usually accompany neonatal Cushing syndrome, so ketoconazole as a medicinal therapy is usually not an option. Metryrapone at an initial dose of 300 mg/m2/day is recommended. The dose may be escalated to as high as 1200 mg/m2/day. In patients with a history of neonatal Cushing, adrenal reserve should be checked. Cushing syndrome in MAS is usually accompanied by hyperthyroidism, which, if not treated, can complicate and worsen the clinical course.

Unresolved questions

While bisphosphonates are usually effective in relieving FD-related pain, whether or not the treatment of FD with bisphosphonates changes the natural history of the disease remains an open question. The most recent, and strongest data to date, suggests that they do not [54]. Ongoing placebo controlled trials in the US and Europe, should help to resolve this open question. More effective treatment for FD is needed. The development of cell based and/or Gsα directed therapies may hold promise.
The best treatment for PP is also not clear. Letrozole, a potent third generation aromatase inhibitor, has recently been shown to be an effective therapy in some girls with MAS [57]. Studies suggest tamoxifen, the estrogen agonist/antagonist, may also be effective [59]. However, all trials for the treatment of PP have been uncontrolled, and this fact, combined with the extreme variability in the clinical course of the disease, makes conclusions about relative efficacy very difficult. A trial with the pure anti-estrogen, fulvestrant, in girls with MAS is underway. Controlled and head-to-head comparison trials are needed to establish the best treatment for PP in MAS.

Support

A list of groups that support patients, families, and clinicians caring for patients with MAS is supplied in Additional file 4.
Written informed consent was obtained from the parents of the patients for publication of this case report and any accompanying images. A copy of the written consent is available for review by the Editor-in-Chief of this journal.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.
Literatur
1.
Zurück zum Zitat McCune DJ: Osteitis fibrosa cystica: the case of a nine-year-old girl who also exhibits precocious puberty, multiple pigmentation of the skin and hyperthyroidism. Am J Dis Child. 1936, 52: 743-744. McCune DJ: Osteitis fibrosa cystica: the case of a nine-year-old girl who also exhibits precocious puberty, multiple pigmentation of the skin and hyperthyroidism. Am J Dis Child. 1936, 52: 743-744.
2.
Zurück zum Zitat Albright F, Butler AM, Hampton AO, Smith P: Syndrome characterized by osteitis fibrosa disseminata, areas, of pigmentation, and endocrine dysfunction, with precocious puberty in females: report of 5 cases. N Engl J Med. 1937, 216: 727-746.CrossRef Albright F, Butler AM, Hampton AO, Smith P: Syndrome characterized by osteitis fibrosa disseminata, areas, of pigmentation, and endocrine dysfunction, with precocious puberty in females: report of 5 cases. N Engl J Med. 1937, 216: 727-746.CrossRef
3.
Zurück zum Zitat Mastorakos G, Mitsiades NS, Doufas AG, Koutras DA: Hyperthyroidism in McCune-Albright syndrome with a review of thyroid abnormalities sixty years after the first report. Thyroid. 1997, 7: 433-439.CrossRefPubMed Mastorakos G, Mitsiades NS, Doufas AG, Koutras DA: Hyperthyroidism in McCune-Albright syndrome with a review of thyroid abnormalities sixty years after the first report. Thyroid. 1997, 7: 433-439.CrossRefPubMed
4.
Zurück zum Zitat Sherman SI, Ladenson PW: Octreotide therapy of growth hormone excess in the McCune-Albright syndrome. Journal of endocrinological investigation. 1992, 15: 185-190.CrossRefPubMed Sherman SI, Ladenson PW: Octreotide therapy of growth hormone excess in the McCune-Albright syndrome. Journal of endocrinological investigation. 1992, 15: 185-190.CrossRefPubMed
5.
Zurück zum Zitat Akintoye SO, Chebli C, Booher S, Feuillan P, Kushner H, Leroith D, Cherman N, Bianco P, Wientroub S, Robey PG, Collins MT: Characterization of gsp-mediated growth hormone excess in the context of McCune-Albright syndrome. J Clin Endocrinol Metab. 2002, 87 (11): 5104-5112. 10.1210/jc.2001-012022.CrossRefPubMed Akintoye SO, Chebli C, Booher S, Feuillan P, Kushner H, Leroith D, Cherman N, Bianco P, Wientroub S, Robey PG, Collins MT: Characterization of gsp-mediated growth hormone excess in the context of McCune-Albright syndrome. J Clin Endocrinol Metab. 2002, 87 (11): 5104-5112. 10.1210/jc.2001-012022.CrossRefPubMed
6.
Zurück zum Zitat Collins MT, Chebli C, Jones J, Kushner H, Consugar M, Rinaldo P, Wientroub S, Bianco P, Robey PG: Renal phosphate wasting in fibrous dysplasia of bone is part of a generalized renal tubular dysfunction similar to that seen in tumor-induced osteomalacia. J Bone Miner Res. 2001, 16: 806-813. 10.1359/jbmr.2001.16.5.806.CrossRefPubMed Collins MT, Chebli C, Jones J, Kushner H, Consugar M, Rinaldo P, Wientroub S, Bianco P, Robey PG: Renal phosphate wasting in fibrous dysplasia of bone is part of a generalized renal tubular dysfunction similar to that seen in tumor-induced osteomalacia. J Bone Miner Res. 2001, 16: 806-813. 10.1359/jbmr.2001.16.5.806.CrossRefPubMed
7.
Zurück zum Zitat Danon M, Crawford JD: The McCune-Albright syndrome. Ergeb Inn Med Kinderheilkd. 1987, 55: 81-115.PubMed Danon M, Crawford JD: The McCune-Albright syndrome. Ergeb Inn Med Kinderheilkd. 1987, 55: 81-115.PubMed
8.
Zurück zum Zitat Diaz A, Danon M, Crawford J: McCune-Albright syndrome and disorders due to activating mutations of GNAS1. J Pediatr Endocrinol Metab. 2007, 20: 853-880.CrossRefPubMed Diaz A, Danon M, Crawford J: McCune-Albright syndrome and disorders due to activating mutations of GNAS1. J Pediatr Endocrinol Metab. 2007, 20: 853-880.CrossRefPubMed
9.
Zurück zum Zitat Kirk JM, Brain CE, Carson DJ, Hyde JC, Grant DB: Cushing's syndrome caused by nodular adrenal hyperplasia in children with McCune-Albright syndrome. The Journal of pediatrics. 1999, 134: 789-792. 10.1016/S0022-3476(99)70302-1.CrossRefPubMed Kirk JM, Brain CE, Carson DJ, Hyde JC, Grant DB: Cushing's syndrome caused by nodular adrenal hyperplasia in children with McCune-Albright syndrome. The Journal of pediatrics. 1999, 134: 789-792. 10.1016/S0022-3476(99)70302-1.CrossRefPubMed
10.
Zurück zum Zitat Shenker A, Weinstein LS, Moran A, Pescovitz OH, Charest NJ, Boney CM, Van Wyk JJ, Merino MJ, Feuillan PP, Spiegel AM: Severe endocrine and nonendocrine manifestations of the McCune-Albright syndrome associated with activating mutations of stimulatory G protein GS. J Pediatr. 1993, 123: 509-518. 10.1016/S0022-3476(05)80943-6.CrossRefPubMed Shenker A, Weinstein LS, Moran A, Pescovitz OH, Charest NJ, Boney CM, Van Wyk JJ, Merino MJ, Feuillan PP, Spiegel AM: Severe endocrine and nonendocrine manifestations of the McCune-Albright syndrome associated with activating mutations of stimulatory G protein GS. J Pediatr. 1993, 123: 509-518. 10.1016/S0022-3476(05)80943-6.CrossRefPubMed
11.
Zurück zum Zitat Lichtenstein LJH: Fibrous dysplasia of bone: a condition affecting one, several or many bones, graver cases of which may present abnormal pigmentation of skin, premature sexual development, hyperthyroidism or still other extraskeletal abnormalities. Arch Path. 1942, 33: 777-816. Lichtenstein LJH: Fibrous dysplasia of bone: a condition affecting one, several or many bones, graver cases of which may present abnormal pigmentation of skin, premature sexual development, hyperthyroidism or still other extraskeletal abnormalities. Arch Path. 1942, 33: 777-816.
12.
Zurück zum Zitat Bianco P, Robey PG, Wientroub S: Fibrous dysplasia. Pediatric Bone: Biology and Disease. Edited by: Glorieux FH, Pettifor J, Juppner H. 2003, New York, NY: Academic Press, Elsevier, 509-539.CrossRef Bianco P, Robey PG, Wientroub S: Fibrous dysplasia. Pediatric Bone: Biology and Disease. Edited by: Glorieux FH, Pettifor J, Juppner H. 2003, New York, NY: Academic Press, Elsevier, 509-539.CrossRef
13.
Zurück zum Zitat Collins MT: Spectrum and natural history of fibrous dysplasia of bone. J Bone Miner Res. 2006, 21 (Suppl 2): P99-P104. 10.1359/jbmr.06s219.CrossRefPubMed Collins MT: Spectrum and natural history of fibrous dysplasia of bone. J Bone Miner Res. 2006, 21 (Suppl 2): P99-P104. 10.1359/jbmr.06s219.CrossRefPubMed
14.
Zurück zum Zitat Collins MT, Bianco P: Fibrous dysplasia. Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism. Edited by: Favus MJ. 2006, Washington, D.C.: American Society for Bone and Mineral Research, 415-418. 6 Collins MT, Bianco P: Fibrous dysplasia. Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism. Edited by: Favus MJ. 2006, Washington, D.C.: American Society for Bone and Mineral Research, 415-418. 6
15.
Zurück zum Zitat Collins MT, Shenker A: McCune-Albright syndrome: new insights. Curr Opin in Endocrinol and Diabetes. 1999, 6: 119-125. 10.1097/00060793-199904000-00006.CrossRef Collins MT, Shenker A: McCune-Albright syndrome: new insights. Curr Opin in Endocrinol and Diabetes. 1999, 6: 119-125. 10.1097/00060793-199904000-00006.CrossRef
16.
Zurück zum Zitat Dorfman HD, Czerniak B: Fibroosseous Lesions. Bone Tumors. Edited by: Dorfman HD, Czerniak B. 1998, St. Louis, MO: Mosby, 441-491. Dorfman HD, Czerniak B: Fibroosseous Lesions. Bone Tumors. Edited by: Dorfman HD, Czerniak B. 1998, St. Louis, MO: Mosby, 441-491.
17.
Zurück zum Zitat Hart ES, Kelly MH, Brillante B, Chen CC, Ziran N, Lee JS, Feuillan P, Leet AI, Kushner H, Robey PG, Collins MT: Onset, progression, and plateau of skeletal lesions in fibrous dysplasia and the relationship to functional outcome. J Bone Miner Res. 2007, 22: 1468-1474. 10.1359/jbmr.070511.CrossRefPubMed Hart ES, Kelly MH, Brillante B, Chen CC, Ziran N, Lee JS, Feuillan P, Leet AI, Kushner H, Robey PG, Collins MT: Onset, progression, and plateau of skeletal lesions in fibrous dysplasia and the relationship to functional outcome. J Bone Miner Res. 2007, 22: 1468-1474. 10.1359/jbmr.070511.CrossRefPubMed
18.
Zurück zum Zitat Leet AI, Chebli C, Kushner H, Chen CC, Kelly MH, Brillante BA, Robey PG, Bianco P, Wientroub S, Collins MT: Fracture incidence in polyostotic fibrous dysplasia and the McCune-Albright syndrome. J Bone Miner Res. 2004, 19: 571-577. 10.1359/JBMR.0301262.CrossRefPubMed Leet AI, Chebli C, Kushner H, Chen CC, Kelly MH, Brillante BA, Robey PG, Bianco P, Wientroub S, Collins MT: Fracture incidence in polyostotic fibrous dysplasia and the McCune-Albright syndrome. J Bone Miner Res. 2004, 19: 571-577. 10.1359/JBMR.0301262.CrossRefPubMed
19.
Zurück zum Zitat Ruggieri P, Sim FH, Bond JR, Unni KK: Malignancies in fibrous dysplasia. Cancer. 1994, 73: 1411-1424. 10.1002/1097-0142(19940301)73:5<1411::AID-CNCR2820730516>3.0.CO;2-T.CrossRefPubMed Ruggieri P, Sim FH, Bond JR, Unni KK: Malignancies in fibrous dysplasia. Cancer. 1994, 73: 1411-1424. 10.1002/1097-0142(19940301)73:5<1411::AID-CNCR2820730516>3.0.CO;2-T.CrossRefPubMed
20.
Zurück zum Zitat Blanco P, Schaeverbeke T, Baillet L, Lequen L, Bannwarth B, Dehais J: Chondrosarcoma in a patient with McCune-Albright syndrome. Report of a case. Rev Rhum Engl Ed. 1999, 66: 177-179.PubMed Blanco P, Schaeverbeke T, Baillet L, Lequen L, Bannwarth B, Dehais J: Chondrosarcoma in a patient with McCune-Albright syndrome. Report of a case. Rev Rhum Engl Ed. 1999, 66: 177-179.PubMed
21.
Zurück zum Zitat Lopez-Ben R, Pitt MJ, Jaffe KA, Siegal GP: Osteosarcoma in a patient with McCune-Albright syndrome and Mazabraud's syndrome. Skeletal Radiol. 1999, 28: 522-526. 10.1007/s002560050556.CrossRefPubMed Lopez-Ben R, Pitt MJ, Jaffe KA, Siegal GP: Osteosarcoma in a patient with McCune-Albright syndrome and Mazabraud's syndrome. Skeletal Radiol. 1999, 28: 522-526. 10.1007/s002560050556.CrossRefPubMed
22.
Zurück zum Zitat Scanlon EF, Burkett FE, Sener SF, Green OC, Traisman HS, Marr TJ, Victor TA, Crist ML: Breast carcinoma in a 11-year-old girl with Albright's syndrome. Breast. 1980, 6: Scanlon EF, Burkett FE, Sener SF, Green OC, Traisman HS, Marr TJ, Victor TA, Crist ML: Breast carcinoma in a 11-year-old girl with Albright's syndrome. Breast. 1980, 6:
23.
Zurück zum Zitat Tanabeu Y, Nakahara S, Mitsuyama S, Ono M, Toyoshima S: Breast Cancer in a Patient with McCune-Albright Syndrome. Breast Cancer. 1998, 5: 175-178.CrossRefPubMed Tanabeu Y, Nakahara S, Mitsuyama S, Ono M, Toyoshima S: Breast Cancer in a Patient with McCune-Albright Syndrome. Breast Cancer. 1998, 5: 175-178.CrossRefPubMed
24.
Zurück zum Zitat Collins MT, Sarlis NJ, Merino MJ, Monroe J, Crawford SE, Krakoff JA, Guthrie LC, Bonat S, Robey PG, Shenker A: Thyroid carcinoma in the McCune-Albright syndrome: contributory role of activating Gs alpha mutations. J Clin Endocrinol Metab. 2003, 88: 4413-4417. 10.1210/jc.2002-021642.CrossRefPubMed Collins MT, Sarlis NJ, Merino MJ, Monroe J, Crawford SE, Krakoff JA, Guthrie LC, Bonat S, Robey PG, Shenker A: Thyroid carcinoma in the McCune-Albright syndrome: contributory role of activating Gs alpha mutations. J Clin Endocrinol Metab. 2003, 88: 4413-4417. 10.1210/jc.2002-021642.CrossRefPubMed
25.
Zurück zum Zitat Weinstein LS, Shenker A, Gejman PV, Merino MJ, Friedman E, Spiegel AM: Activating mutations of the stimulatory G protein in the McCune – Albright syndrome [see comments]. N Engl J Med. 1991, 325: 1688-1695.CrossRefPubMed Weinstein LS, Shenker A, Gejman PV, Merino MJ, Friedman E, Spiegel AM: Activating mutations of the stimulatory G protein in the McCune – Albright syndrome [see comments]. N Engl J Med. 1991, 325: 1688-1695.CrossRefPubMed
26.
Zurück zum Zitat Schwindinger WF, Francomano CA, Levine MA: Identification of a mutation in the gene encoding the alpha subunit of the stimulatory G protein of adenylyl cyclase in McCune-Albright syndrome. Proc Natl Acad Sci USA. 1992, 89: 5152-5156. 10.1073/pnas.89.11.5152.PubMedCentralCrossRefPubMed Schwindinger WF, Francomano CA, Levine MA: Identification of a mutation in the gene encoding the alpha subunit of the stimulatory G protein of adenylyl cyclase in McCune-Albright syndrome. Proc Natl Acad Sci USA. 1992, 89: 5152-5156. 10.1073/pnas.89.11.5152.PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Bianco P, Riminucci M, Majolagbe A, Kuznetsov SA, Collins MT, Mankani MH, Corsi A, Bone HG, Wientroub S, Spiegel AM, Fisher LW, Robey PG: Mutations of the GNAS1 gene, stromal cell dysfunction, and osteomalacic changes in non-McCune-Albright fibrous dysplasia of bone. J Bone Miner Res. 2000, 15: 120-128. 10.1359/jbmr.2000.15.1.120.CrossRefPubMed Bianco P, Riminucci M, Majolagbe A, Kuznetsov SA, Collins MT, Mankani MH, Corsi A, Bone HG, Wientroub S, Spiegel AM, Fisher LW, Robey PG: Mutations of the GNAS1 gene, stromal cell dysfunction, and osteomalacic changes in non-McCune-Albright fibrous dysplasia of bone. J Bone Miner Res. 2000, 15: 120-128. 10.1359/jbmr.2000.15.1.120.CrossRefPubMed
28.
Zurück zum Zitat Idowu BD, Al-Adnani M, O'Donnell P, Yu L, Odell E, Diss T, Gale RE, Flanagan AM: A sensitive mutation-specific screening technique for GNAS1 mutations in cases of fibrous dysplasia: the first report of a codon 227 mutation in bone. Histopathology. 2007, 50: 691-704. 10.1111/j.1365-2559.2007.02676.x.CrossRefPubMed Idowu BD, Al-Adnani M, O'Donnell P, Yu L, Odell E, Diss T, Gale RE, Flanagan AM: A sensitive mutation-specific screening technique for GNAS1 mutations in cases of fibrous dysplasia: the first report of a codon 227 mutation in bone. Histopathology. 2007, 50: 691-704. 10.1111/j.1365-2559.2007.02676.x.CrossRefPubMed
29.
Zurück zum Zitat Riminucci M, Saggio I, Robey PG, Bianco P: Fibrous dysplasia as a stem cell disease. J Bone Miner Res. 2006, 21 (Suppl 2): P125-131. 10.1359/jbmr.06s224.CrossRefPubMed Riminucci M, Saggio I, Robey PG, Bianco P: Fibrous dysplasia as a stem cell disease. J Bone Miner Res. 2006, 21 (Suppl 2): P125-131. 10.1359/jbmr.06s224.CrossRefPubMed
30.
Zurück zum Zitat Collins MT, Kushner H, Reynolds JC, Chebli C, Kelly MH, Gupta A, Brillante B, Leet AI, Riminucci M, Robey PG, Bianco P, Wientroub S, Chen CC: An instrument to measure skeletal burden and predict functional outcome in fibrous dysplasia of bone. J Bone Miner Res. 2005, 20: 219-226. 10.1359/JBMR.041111.CrossRefPubMed Collins MT, Kushner H, Reynolds JC, Chebli C, Kelly MH, Gupta A, Brillante B, Leet AI, Riminucci M, Robey PG, Bianco P, Wientroub S, Chen CC: An instrument to measure skeletal burden and predict functional outcome in fibrous dysplasia of bone. J Bone Miner Res. 2005, 20: 219-226. 10.1359/JBMR.041111.CrossRefPubMed
31.
Zurück zum Zitat Ippolito E, Bray EW, Corsi A, De Maio F, Exner UG, Robey PG, Grill F, Lala R, Massobrio M, Pinggera O, Riminucci M, Snela S, Zambakidis C, Bianco P: Natural history and treatment of fibrous dysplasia of bone: a multicenter clinicopathologic study promoted by the European Pediatric Orthopaedic Society. Journal of pediatric orthopaedics. 2003, 12: 155-177. 10.1097/00009957-200305000-00001. Ippolito E, Bray EW, Corsi A, De Maio F, Exner UG, Robey PG, Grill F, Lala R, Massobrio M, Pinggera O, Riminucci M, Snela S, Zambakidis C, Bianco P: Natural history and treatment of fibrous dysplasia of bone: a multicenter clinicopathologic study promoted by the European Pediatric Orthopaedic Society. Journal of pediatric orthopaedics. 2003, 12: 155-177. 10.1097/00009957-200305000-00001.
32.
Zurück zum Zitat Kelly MH, Brillante B, Collins MT: Pain in fibrous dysplasia of bone: age-related changes and the anatomical distribution of skeletal lesions. Osteoporos Int. 2007 Kelly MH, Brillante B, Collins MT: Pain in fibrous dysplasia of bone: age-related changes and the anatomical distribution of skeletal lesions. Osteoporos Int. 2007
33.
Zurück zum Zitat Riminucci M, Liu B, Corsi A, Shenker A, Spiegel AM, Robey PG, Bianco P: The histopathology of fibrous dysplasia of bone in patients with activating mutations of the Gs alpha gene: site-specific patterns and recurrent histological hallmarks. The Journal of pathology. 1999, 187: 249-258. 10.1002/(SICI)1096-9896(199901)187:2<249::AID-PATH222>3.0.CO;2-J.CrossRefPubMed Riminucci M, Liu B, Corsi A, Shenker A, Spiegel AM, Robey PG, Bianco P: The histopathology of fibrous dysplasia of bone in patients with activating mutations of the Gs alpha gene: site-specific patterns and recurrent histological hallmarks. The Journal of pathology. 1999, 187: 249-258. 10.1002/(SICI)1096-9896(199901)187:2<249::AID-PATH222>3.0.CO;2-J.CrossRefPubMed
34.
Zurück zum Zitat Corsi A, Collins MT, Riminucci M, Howell PG, Boyde A, Robey PG, Bianco P: Osteomalacic and hyperparathyroid changes in fibrous dysplasia of bone: core biopsy studies and clinical correlations. J Bone Miner Res. 2003, 18: 1235-1246. 10.1359/jbmr.2003.18.7.1235.CrossRefPubMed Corsi A, Collins MT, Riminucci M, Howell PG, Boyde A, Robey PG, Bianco P: Osteomalacic and hyperparathyroid changes in fibrous dysplasia of bone: core biopsy studies and clinical correlations. J Bone Miner Res. 2003, 18: 1235-1246. 10.1359/jbmr.2003.18.7.1235.CrossRefPubMed
35.
Zurück zum Zitat Hannon TS, Noonan K, Steinmetz R, Eugster EA, Levine MA, Pescovitz OH: Is McCune-Albright syndrome overlooked in subjects with fibrous dysplasia of bone?. J Pediatr. 2003, 142: 532-538. 10.1067/mpd.2003.153.CrossRefPubMed Hannon TS, Noonan K, Steinmetz R, Eugster EA, Levine MA, Pescovitz OH: Is McCune-Albright syndrome overlooked in subjects with fibrous dysplasia of bone?. J Pediatr. 2003, 142: 532-538. 10.1067/mpd.2003.153.CrossRefPubMed
38.
Zurück zum Zitat Byard RW: Forensic considerations in cases of neurofibromatosis – an overview. Journal of forensic sciences. 2007, 52: 1164-1170. 10.1111/j.1556-4029.2007.00512.x.CrossRefPubMed Byard RW: Forensic considerations in cases of neurofibromatosis – an overview. Journal of forensic sciences. 2007, 52: 1164-1170. 10.1111/j.1556-4029.2007.00512.x.CrossRefPubMed
39.
Zurück zum Zitat Theos A, Korf BR: Pathophysiology of neurofibromatosis type 1. Annals of internal medicine. 2006, 144: 842-849.CrossRefPubMed Theos A, Korf BR: Pathophysiology of neurofibromatosis type 1. Annals of internal medicine. 2006, 144: 842-849.CrossRefPubMed
40.
Zurück zum Zitat Lee JS, FitzGibbon E, Butman JA, Dufresne CR, Kushner H, Wientroub S, Robey PG, Collins MT: Normal vision despite narrowing of the optic canal in fibrous dysplasia. N Engl J Med. 2002, 347: 1670-1676. 10.1056/NEJMoa020742.CrossRefPubMed Lee JS, FitzGibbon E, Butman JA, Dufresne CR, Kushner H, Wientroub S, Robey PG, Collins MT: Normal vision despite narrowing of the optic canal in fibrous dysplasia. N Engl J Med. 2002, 347: 1670-1676. 10.1056/NEJMoa020742.CrossRefPubMed
41.
Zurück zum Zitat Cutler CM, Lee JS, Butman JA, FitzGibbon EJ, Kelly MH, Brillante BA, Feuillan P, Robey PG, DuFresne CR, Collins MT: Long-term outcome of optic nerve encasement and optic nerve decompression in patients with fibrous dysplasia: risk factors for blindness and safety of observation. Neurosurgery. 2006, 59: 1011-1017. discussion 1017–1018PubMed Cutler CM, Lee JS, Butman JA, FitzGibbon EJ, Kelly MH, Brillante BA, Feuillan P, Robey PG, DuFresne CR, Collins MT: Long-term outcome of optic nerve encasement and optic nerve decompression in patients with fibrous dysplasia: risk factors for blindness and safety of observation. Neurosurgery. 2006, 59: 1011-1017. discussion 1017–1018PubMed
42.
Zurück zum Zitat Feuillan PP, Shawker T, Rose SR, Jones J, Jeevanram RK, Nisula BC: Thyroid abnormalities in the McCune-Albright syndrome: ultrasonography and hormone studies. J Clin Endocrinol Metab. 1990, 71: 1596-1601.CrossRefPubMed Feuillan PP, Shawker T, Rose SR, Jones J, Jeevanram RK, Nisula BC: Thyroid abnormalities in the McCune-Albright syndrome: ultrasonography and hormone studies. J Clin Endocrinol Metab. 1990, 71: 1596-1601.CrossRefPubMed
43.
Zurück zum Zitat 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: FGF-23 in fibrous dysplasia of bone and its relationship to renal phosphate wasting. J Clin Invest. 2003, 112: 683-692.PubMedCentralCrossRefPubMed 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: FGF-23 in fibrous dysplasia of bone and its relationship to renal phosphate wasting. J Clin Invest. 2003, 112: 683-692.PubMedCentralCrossRefPubMed
44.
Zurück zum Zitat Terpstra L, Rauch F, Plotkin H, Travers R, Glorieux FH: Bone mineralization in polyostotic fibrous dysplasia: histomorphometric analysis. J Bone Miner Res. 2002, 17: 1949-1953. 10.1359/jbmr.2002.17.11.1949.CrossRefPubMed Terpstra L, Rauch F, Plotkin H, Travers R, Glorieux FH: Bone mineralization in polyostotic fibrous dysplasia: histomorphometric analysis. J Bone Miner Res. 2002, 17: 1949-1953. 10.1359/jbmr.2002.17.11.1949.CrossRefPubMed
45.
Zurück zum Zitat Glorieux FH, Rauch F: Medical therapy of children with fibrous dysplasia. J Bone Miner Res. 2006, 21 (Suppl 2): P110-113. 10.1359/jbmr.06s221.CrossRefPubMed Glorieux FH, Rauch F: Medical therapy of children with fibrous dysplasia. J Bone Miner Res. 2006, 21 (Suppl 2): P110-113. 10.1359/jbmr.06s221.CrossRefPubMed
46.
Zurück zum Zitat Hammami MM, Hussain SS, Vencer LJ, Butt A, al-Zahrani A: Primary hyperparathyroidism-associated polyostotic fibrous dysplasia: absence of McCune-Albright syndrome mutations. J Endocrinol Invest. 1997, 20: 552-558.CrossRefPubMed Hammami MM, Hussain SS, Vencer LJ, Butt A, al-Zahrani A: Primary hyperparathyroidism-associated polyostotic fibrous dysplasia: absence of McCune-Albright syndrome mutations. J Endocrinol Invest. 1997, 20: 552-558.CrossRefPubMed
47.
Zurück zum Zitat Barone A, Giusti A, Pioli G, Girasole G, Razzano M, Pizzonia M, Palummeri E, Bianchi G: Secondary hyperparathyroidism due to hypovitaminosis D affects bone mineral density response to alendronate in elderly women with osteoporosis: a randomized controlled trial. Journal of the American Geriatrics Society. 2007, 55: 752-757. 10.1111/j.1532-5415.2007.01161.x.CrossRefPubMed Barone A, Giusti A, Pioli G, Girasole G, Razzano M, Pizzonia M, Palummeri E, Bianchi G: Secondary hyperparathyroidism due to hypovitaminosis D affects bone mineral density response to alendronate in elderly women with osteoporosis: a randomized controlled trial. Journal of the American Geriatrics Society. 2007, 55: 752-757. 10.1111/j.1532-5415.2007.01161.x.CrossRefPubMed
48.
Zurück zum Zitat Giusti A, Barone A, Razzano M, Pizzonia M, Oliveri M, Palummeri E, Pioli G: High prevalence of secondary hyperparathyroidism due to hypovitaminosis D in hospitalized elderly with and without hip fracture. Journal of endocrinological investigation. 2006, 29: 809-813.CrossRefPubMed Giusti A, Barone A, Razzano M, Pizzonia M, Oliveri M, Palummeri E, Pioli G: High prevalence of secondary hyperparathyroidism due to hypovitaminosis D in hospitalized elderly with and without hip fracture. Journal of endocrinological investigation. 2006, 29: 809-813.CrossRefPubMed
49.
Zurück zum Zitat Reginster JY: The high prevalence of inadequate serum vitamin D levels and implications for bone health. Current medical research and opinion. 2005, 21: 579-586. 10.1185/030079905X41435.CrossRefPubMed Reginster JY: The high prevalence of inadequate serum vitamin D levels and implications for bone health. Current medical research and opinion. 2005, 21: 579-586. 10.1185/030079905X41435.CrossRefPubMed
50.
Zurück zum Zitat Hashemipour S, Larijani B, Adibi H, Sedaghat M, Pajouhi M, Bastan-Hagh MH, Soltani A, Javadi E, Shafaei AR, Baradar-Jalili R, Hossein-Nezhad A: The status of biochemical parameters in varying degrees of vitamin D deficiency. Journal of bone and mineral metabolism. 2006, 24: 213-218. 10.1007/s00774-005-0674-8.CrossRefPubMed Hashemipour S, Larijani B, Adibi H, Sedaghat M, Pajouhi M, Bastan-Hagh MH, Soltani A, Javadi E, Shafaei AR, Baradar-Jalili R, Hossein-Nezhad A: The status of biochemical parameters in varying degrees of vitamin D deficiency. Journal of bone and mineral metabolism. 2006, 24: 213-218. 10.1007/s00774-005-0674-8.CrossRefPubMed
51.
Zurück zum Zitat Stanton RP: Surgery for fibrous dysplasia. J Bone Miner Res. 2006, 21 (Suppl 2): P105-109. 10.1359/jbmr.06s220.CrossRefPubMed Stanton RP: Surgery for fibrous dysplasia. J Bone Miner Res. 2006, 21 (Suppl 2): P105-109. 10.1359/jbmr.06s220.CrossRefPubMed
52.
Zurück zum Zitat Chapurlat RD: Medical therapy in adults with fibrous dysplasia of bone. J Bone Miner Res. 2006, 21 (Suppl 2): P114-119. 10.1359/jbmr.06s222.CrossRefPubMed Chapurlat RD: Medical therapy in adults with fibrous dysplasia of bone. J Bone Miner Res. 2006, 21 (Suppl 2): P114-119. 10.1359/jbmr.06s222.CrossRefPubMed
53.
Zurück zum Zitat Liens D, Delmas PD, Meunier PJ: Long-term effects of intravenous pamidronate in fibrous dysplasia of bone. Lancet. 1994, 343: 953-954. 10.1016/S0140-6736(94)90069-8.CrossRefPubMed Liens D, Delmas PD, Meunier PJ: Long-term effects of intravenous pamidronate in fibrous dysplasia of bone. Lancet. 1994, 343: 953-954. 10.1016/S0140-6736(94)90069-8.CrossRefPubMed
54.
Zurück zum Zitat Plotkin H, Rauch F, Zeitlin L, Munns C, Travers R, Glorieux FH: Effect of pamidronate treatment in children with polyostotic fibrous dysplasia of bone. J Clin Endocrinol Metab. 2003, 88: 4569-4575. 10.1210/jc.2003-030050.CrossRefPubMed Plotkin H, Rauch F, Zeitlin L, Munns C, Travers R, Glorieux FH: Effect of pamidronate treatment in children with polyostotic fibrous dysplasia of bone. J Clin Endocrinol Metab. 2003, 88: 4569-4575. 10.1210/jc.2003-030050.CrossRefPubMed
55.
Zurück zum Zitat Chan B, Zacharin M: Pamidronate treatment of polyostotic fibrous dysplasia: failure to prevent expansion of dysplastic lesions during childhood. J Pediatr Endocrinol Metab. 2006, 19: 75-80.PubMed Chan B, Zacharin M: Pamidronate treatment of polyostotic fibrous dysplasia: failure to prevent expansion of dysplastic lesions during childhood. J Pediatr Endocrinol Metab. 2006, 19: 75-80.PubMed
56.
Zurück zum Zitat Leet AI, Magur E, Lee JS, Wientroub S, Robey PG, Collins MT: Fibrous dysplasia in the spine: prevalence of lesions and association with scoliosis. J Bone Joint Surg Am. 2004, 86-A (3): 531-537.PubMed Leet AI, Magur E, Lee JS, Wientroub S, Robey PG, Collins MT: Fibrous dysplasia in the spine: prevalence of lesions and association with scoliosis. J Bone Joint Surg Am. 2004, 86-A (3): 531-537.PubMed
57.
Zurück zum Zitat Feuillan P, Calis K, Hill S, Shawker T, Robey PG, Collins MT: Letrozole Treatment of Precocious Puberty in Girls with the McCune-Albright Syndrome: A Pilot Study. J Clin Endocrinol Metab. 2007, 92: 2100-2106. 10.1210/jc.2006-2350.CrossRefPubMed Feuillan P, Calis K, Hill S, Shawker T, Robey PG, Collins MT: Letrozole Treatment of Precocious Puberty in Girls with the McCune-Albright Syndrome: A Pilot Study. J Clin Endocrinol Metab. 2007, 92: 2100-2106. 10.1210/jc.2006-2350.CrossRefPubMed
58.
Zurück zum Zitat Feuillan PP: Treatment of sexual precocity in girls with the McCune-Albright syndrome. Sexual precocity: etiology, diagnosis and management. Edited by: Grave GD, Cutler GB. 1993, New York: Raven Press, 243-251. Feuillan PP: Treatment of sexual precocity in girls with the McCune-Albright syndrome. Sexual precocity: etiology, diagnosis and management. Edited by: Grave GD, Cutler GB. 1993, New York: Raven Press, 243-251.
59.
Zurück zum Zitat Eugster EA, Rubin SD, Reiter EO, Plourde P, Jou HC, Pescovitz OH: Tamoxifen treatment for precocious puberty in McCune-Albright syndrome: a multicenter trial. The Journal of pediatrics. 2003, 143: 60-66. 10.1016/S0022-3476(03)00128-8.CrossRefPubMed Eugster EA, Rubin SD, Reiter EO, Plourde P, Jou HC, Pescovitz OH: Tamoxifen treatment for precocious puberty in McCune-Albright syndrome: a multicenter trial. The Journal of pediatrics. 2003, 143: 60-66. 10.1016/S0022-3476(03)00128-8.CrossRefPubMed
60.
Zurück zum Zitat Congedo V, Celi FS: Thyroid disease in patients with McCune-Albright syndrome. Pediatr Endocrinol Rev. 2007, 4 (Suppl 4): 429-433.PubMed Congedo V, Celi FS: Thyroid disease in patients with McCune-Albright syndrome. Pediatr Endocrinol Rev. 2007, 4 (Suppl 4): 429-433.PubMed
61.
Zurück zum Zitat Akintoye SO, Kelly MH, Brillante B, Cherman N, Turner S, Butman JA, Robey PG, Collins MT: Pegvisomant for the treatment of gsp-mediated growth hormone excess in patients with McCune-Albright syndrome. J Clin Endocrinol Metab. 2006, 91: 2960-2966. 10.1210/jc.2005-2661.CrossRefPubMed Akintoye SO, Kelly MH, Brillante B, Cherman N, Turner S, Butman JA, Robey PG, Collins MT: Pegvisomant for the treatment of gsp-mediated growth hormone excess in patients with McCune-Albright syndrome. J Clin Endocrinol Metab. 2006, 91: 2960-2966. 10.1210/jc.2005-2661.CrossRefPubMed
62.
Zurück zum Zitat Chanson P, Salenave S, Orcel P: McCune-Albright syndrome in adulthood. Pediatr Endocrinol Rev. 2007, 4 (Suppl 4): 453-462.PubMed Chanson P, Salenave S, Orcel P: McCune-Albright syndrome in adulthood. Pediatr Endocrinol Rev. 2007, 4 (Suppl 4): 453-462.PubMed
63.
Zurück zum Zitat Galland F, Kamenicky P, Affres H, Reznik Y, Pontvert D, Le Bouc Y, Young J, Chanson P: McCune-Albright syndrome and acromegaly: effects of hypothalamopituitary radiotherapy and/or pegvisomant in somatostatin analog-resistant patients. J Clin Endocrinol Metab. 2006, 91: 4957-4961. 10.1210/jc.2006-0561.CrossRefPubMed Galland F, Kamenicky P, Affres H, Reznik Y, Pontvert D, Le Bouc Y, Young J, Chanson P: McCune-Albright syndrome and acromegaly: effects of hypothalamopituitary radiotherapy and/or pegvisomant in somatostatin analog-resistant patients. J Clin Endocrinol Metab. 2006, 91: 4957-4961. 10.1210/jc.2006-0561.CrossRefPubMed
64.
Zurück zum Zitat Ehrig U, Wilson DR: Fibrous dysplasia of bone and primary hyperparathyroidism. Ann Intern Med. 1972, 77: 234-238.CrossRefPubMed Ehrig U, Wilson DR: Fibrous dysplasia of bone and primary hyperparathyroidism. Ann Intern Med. 1972, 77: 234-238.CrossRefPubMed
65.
Zurück zum Zitat Leslie WD, Reinhold C, Rosenthall L, Tau C, Glorieux FH: Panostotic fibrous dysplasia. A new craniotubular dysplasia. Clinical nuclear medicine. 1992, 17: 556-560. 10.1097/00003072-199207000-00005.CrossRefPubMed Leslie WD, Reinhold C, Rosenthall L, Tau C, Glorieux FH: Panostotic fibrous dysplasia. A new craniotubular dysplasia. Clinical nuclear medicine. 1992, 17: 556-560. 10.1097/00003072-199207000-00005.CrossRefPubMed
66.
Zurück zum Zitat Feuillan PP: McCune-Albright syndrome. Curr Ther Endocrinol Metab. 1997, 6: 235-239.PubMed Feuillan PP: McCune-Albright syndrome. Curr Ther Endocrinol Metab. 1997, 6: 235-239.PubMed
67.
Zurück zum Zitat Defilippi C, Chiappetta D, Marzari D, Mussa A, Lala R: Image diagnosis in McCune-Albright syndrome. J Pediatr Endocrinol Metab. 2006, 19 (Suppl 2): 561-570.PubMed Defilippi C, Chiappetta D, Marzari D, Mussa A, Lala R: Image diagnosis in McCune-Albright syndrome. J Pediatr Endocrinol Metab. 2006, 19 (Suppl 2): 561-570.PubMed
68.
Zurück zum Zitat Silva ES, Lumbroso S, Medina M, Gillerot Y, Sultan C, Sokal EM: Demonstration of McCune-Albright mutations in the liver of children with high gammaGT progressive cholestasis. Journal of hepatology. 2000, 32: 154-158. 10.1016/S0168-8278(00)80202-0.CrossRefPubMed Silva ES, Lumbroso S, Medina M, Gillerot Y, Sultan C, Sokal EM: Demonstration of McCune-Albright mutations in the liver of children with high gammaGT progressive cholestasis. Journal of hepatology. 2000, 32: 154-158. 10.1016/S0168-8278(00)80202-0.CrossRefPubMed
70.
Zurück zum Zitat Schwartz RA, Spicer MS, Leevy CB, Ticker JB, Lambert WC: Cutaneous fibrous dysplasia: an incomplete form of the McCune-Albright syndrome. Dermatology (Basel, Switzerland). 1996, 192: 258-261.CrossRef Schwartz RA, Spicer MS, Leevy CB, Ticker JB, Lambert WC: Cutaneous fibrous dysplasia: an incomplete form of the McCune-Albright syndrome. Dermatology (Basel, Switzerland). 1996, 192: 258-261.CrossRef
71.
Zurück zum Zitat Pierini AM, Ortonne JP, Floret D: [Cutaneous manifestations of McCune-Albright syndrome: report of a case (author's transl)]. Annales de dermatologie et de venereologie. 1981, 108: 969-976.PubMed Pierini AM, Ortonne JP, Floret D: [Cutaneous manifestations of McCune-Albright syndrome: report of a case (author's transl)]. Annales de dermatologie et de venereologie. 1981, 108: 969-976.PubMed
Metadaten
Titel
McCune-Albright syndrome
verfasst von
Claudia E Dumitrescu
Michael T Collins
Publikationsdatum
01.12.2008
Verlag
BioMed Central
Erschienen in
Orphanet Journal of Rare Diseases / Ausgabe 1/2008
Elektronische ISSN: 1750-1172
DOI
https://doi.org/10.1186/1750-1172-3-12

Weitere Artikel der Ausgabe 1/2008

Orphanet Journal of Rare Diseases 1/2008 Zur Ausgabe