ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

NF2-related schwannomatosis (formerly neurofibromatosis type 2)

NF2-related schwannomatosis (formerly neurofibromatosis type 2)
Literature review current through: Jan 2024.
This topic last updated: Nov 02, 2023.

INTRODUCTION — NF2-related schwannomatosis (formerly neurofibromatosis type 2), hereafter referred to as "NF2" [1], is a dominantly inherited syndrome that predisposes individuals to bilateral vestibular schwannomas as well as multiple other tumors of the nervous system [2-4]. NF2 is caused by pathogenic variants in the NF2, moesin-ezrin-radixin like (MERLIN) tumor suppressor (NF2) gene, which produces merlin, a tumor suppressor.

The molecular pathogenesis, clinical features, diagnosis, and patient management of NF2 are reviewed here. Neurofibromatosis type 1 (NF1; von Recklinghausen disease) and other schwannomatoses are discussed separately. (See "Neurofibromatosis type 1 (NF1): Pathogenesis, clinical features, and diagnosis" and "Schwannomatoses related to genetic variants other than NF2".)

TERMINOLOGY — In 2022, the nomenclature for NF2 and schwannomatosis was updated to recognize NF2 and schwannomatosis as parts of a spectrum of schwannoma predisposition syndromes, which are defined in many cases by pathogenic variants in one of several genes on chromosome 22 [1]:

NF2-related schwannomatosis (and shortened to NF2 in this topic and others for brevity), formerly neurofibromatosis type 2

SMARCB1-related schwannomatosis

LZTR1-related schwannomatosis

Other schwannomatoses, including those related to loss of heterozygosity of chromosome 22q

MOLECULAR PATHOGENESIS

NF2 gene — NF2 is consistently linked with abnormalities of the NF2 gene, which is located on chromosome 22 [5]. The NF2 gene produces merlin, also known as schwannomin, a cell membrane-related protein that acts as a tumor suppressor [6,7].

Detailed molecular testing will identify an abnormality in NF2 in over 93 percent of families with multiple members affected with NF2 [8]. Individuals with NF2 may inherit an abnormal NF2 allele from a parent. Alternatively, a de novo variant may arise after fertilization, resulting in a mosaic expression of two cell lines. For individuals thought to harbor a de novo variant, somatic mosaicism may prevent the molecular diagnosis from being established unless tumor tissue is analyzed [8-12]. A study of over 1000 patients with de novo NF2 found that more than 50 percent of such cases are potentially due to mosaicism [13].

The development of schwannomas and other tumors requires inactivation of both NF2 alleles, since tumors only develop in cells that have lost function of their normal NF2 allele. The "second hit" usually occurs through loss of the entire healthy NF2 gene and most or all of chromosome 22, although a point pathogenic variant or promoter methylation may also occur. Loss of the normal copy may also occur by mitotic recombination, where the mutated copy is doubled up without loss of chromosomal material [14].

Genotype-phenotype correlation — NF2 is fully penetrant, meaning that any individual who carries a germline pathogenic NF2 variant will develop the clinical disorder [1]. Within a family that includes multiple members with NF2, the phenotypic expression and natural history of the disease are similar. However, significant differences have been observed among families harboring different abnormalities of NF2.

Frameshift or nonsense variants cause truncated protein expression, which has been associated with more severe manifestations of NF2; missense or inframe deletions have been associated with milder manifestations of disease [15]. In an analysis of 268 patients in whom the NF2 abnormality was characterized, the presence of a truncated protein was associated with younger age at diagnosis and a higher prevalence of meningiomas, spinal tumors, and tumors of cranial nerves other than VIII [16]. Patients with variants causing truncated protein also had vestibular schwannomas at a younger age and had more cutaneous tumors. There is also a positional effect, with variants in the later parts of the gene (especially exons 14 and 15) being associated with milder disease and fewer meningiomas [17].

There is also evidence that these differences have marked effects on life expectancy, with missense variants being associated with the best life expectancy and truncating variants in exons 2 to 13 having the poorest survival [18]. A genetic severity score can be derived from the variant identified, which predicts the clinical severity of NF2 disease (table 1) [18,19].

EPIDEMIOLOGY — The estimated incidence of NF2 derived from population-based studies in England and Finland is as high as 1 in 25,000 [20-23].

CLINICAL FEATURES — NF2 is an autosomal dominant schwannoma predisposition syndrome that affects the nervous system as well as eyes and skin. In addition to schwannomas and other nervous system tumors, most patients have one or more ophthalmic and cutaneous manifestations.

Age of onset and clinical spectrum — Patients typically present around 20 to 25 years of age [15]. Initial or presenting symptoms vary by age at presentation.

Children diagnosed with NF2 often have an atypical, but more severe, presentation [24]. In childhood, patients most commonly present with visual/eye problems, hearing loss, weakness, pain, mononeuropathy, cutaneous tumors, and/or seizures [15,25,26]. In adults, hearing loss and tinnitus are the presenting symptoms in over one-half of patients.

The most frequent clinical features of NF2 include:

Neurologic lesions [27-29]

Bilateral vestibular schwannomas, generally developing by 30 years of age – 90 to 95 percent

Schwannomas of other cranial nerves – 24 to 51 percent

Intracranial meningiomas – 45 to 77 percent [17]

Spinal tumors (both intramedullary and extramedullary) – 63 to 90 percent

Peripheral neuropathy – Up to 66 percent

Eye lesions [30-32]

Cataracts – 60 to 81 percent

Epiretinal membranes – 12 to 40 percent

Retinal hamartomas – 6 to 22 percent

Skin lesions [27,29,33,34]

Cutaneous tumors – 59 to 68 percent

Skin plaques – 41 to 48 percent

Subcutaneous tumors – 43 to 48 percent

Vestibular schwannomas — Vestibular schwannomas in NF2 are typically bilateral and cause tinnitus, hearing loss, and balance dysfunction (image 1) [3]. The onset of hearing loss is usually gradual and progressive, leading to deafness, although sudden hearing loss can occur. NF2-related vestibular schwannomas are usually multifocal and occur equally on the superior and inferior vestibular nerve [35].

The mechanism of hearing loss from vestibular schwannomas in NF2 is not well understood, and there is poor correlation between tumor size and/or growth rate and degree of hearing loss [36-39]. One proposed mechanism is the accumulation of intralabyrinthine protein as a result of cochlear aperture obstruction, which can be visualized by high-resolution magnetic resonance imaging (MRI) [40]. If left untreated, vestibular schwannomas can extend medially and cause brainstem compression and hydrocephalus. (See "Vestibular schwannoma (acoustic neuroma)", section on 'Clinical presentation'.)

Almost all vestibular schwannomas in patients with NF2 are histopathologically benign. However, they are an important cause of morbidity because of their anatomic location and bilaterality.

Meningiomas — Approximately one-half of individuals with NF2 have meningiomas, and multiple meningiomas are often present [41]. The incidence increases with age, and lifetime risk may be as high as 75 percent [29]. Most meningiomas are intracranial, although spinal meningiomas are also seen. (See "Epidemiology, pathology, clinical features, and diagnosis of meningioma", section on 'Clinical presentation' and "Spinal cord tumors", section on 'Meningioma'.)

Patients with NF2 tend to develop meningiomas at an earlier age than those with sporadic meningiomas. When patients are diagnosed with meningioma during childhood, approximately 20 percent will be found to have NF2 [24,42]. Histopathologically, the meningiomas seen in patients with NF2 are more frequently atypical or anaplastic compared with sporadic tumors [43,44].

Spinal tumors — Most individuals with NF2 eventually develop a spinal tumor, which can cause debilitating pain, muscle weakness, or paresthesias [3,45,46].

Schwannomas are most common (image 2). These arise from the dorsal root and can take on a characteristic "dumbbell" shape. (See "Spinal cord tumors", section on 'Presentation' and "Intradural nerve sheath tumors", section on 'Clinical presentation'.)

Meningiomas can also arise on the spinal cord in the extramedullary space. (See "Spinal cord tumors", section on 'Meningioma'.)

Intramedullary tumors, particularly ependymomas (image 3), are seen in 5 to 33 percent of cases. (See "Spinal cord tumors", section on 'Ependymomas'.)

Neuropathies — Neuropathies can be manifested in a variety of ways in individuals with NF2 [2,47]. Some people develop a mononeuropathy, often involving the facial nerve, which can precede the development of other NF2 manifestations. In 3 to 5 percent of adults, a more severe polyneuropathy not directly related to tumors is seen, and this can progress to severe muscle wasting [2,3,48].

Ophthalmic manifestations — Patients with NF2 may have visual impairment due to cataracts, optic nerve meningiomas, retinal hamartomas, and epiretinal membrane [2,3,26,32,49]. Cataracts are seen in 60 to 80 percent of patients and typically are manifested as posterior subcapsular lenticular opacities. Ophthalmic manifestations are more severe with increased genetic severity score [50]. (See 'Genotype-phenotype correlation' above.)

Development of cataracts and other ocular manifestations can lead to visual abnormalities in early childhood in those with NF2 [26]. In addition, particular attention is required to differentiate optic nerve meningiomas or hamartomas from retinoblastoma. (See "Retinoblastoma: Clinical presentation, evaluation, and diagnosis", section on 'Clinical presentation'.)

Cutaneous manifestations — Approximately 70 percent of patients with NF2 have cutaneous manifestations, although only 10 percent have more than 10 skin tumors [2]. These skin lesions can take several forms:

Plaque-like lesions, which are intracutaneous and slightly raised [12]. These may be more pigmented than the surrounding skin and may be associated with increased hair.

Subcutaneous nodules can often be palpated, sometimes along the course of a peripheral nerve. These tumors represent a swelling of the nerve.

Intracutaneous tumors, similar to those observed in patients with neurofibromatosis type 1 (NF1), are also seen occasionally. These tumors generally represent schwannomas rather than neurofibromas.

DIAGNOSIS — The diagnosis of NF2 is based upon the presence of characteristic clinical and molecular genetic features. Genetic testing is recommended in all patients with suspected schwannomatosis predisposition syndromes but is not required for the diagnosis of NF2 in patients who fulfill clinical criteria. Genetic testing is particularly helpful in younger patients who do not meet diagnostic criteria without genetic data. Genetic testing is also important for first-degree relatives of an individual with NF2, who can be diagnosed with NF2 based on an NF2 pathogenic variant, even in the absence of clinical features.

Clinical and radiologic evaluation — For individuals suspected of having NF2, the initial evaluation should include a detailed clinical and family history, neurologic examination, careful cutaneous and eye examinations, and contrast-enhanced MRI of the brain and whole spine [2].

Skin examination – A full skin examination should be performed to assess for cutaneous tumor burden. Suspicious lesions should be palpated. (See 'Cutaneous manifestations' above.)

Eye examination – All patients should have a slit lamp examination and ophthalmoscopy, ideally by an ophthalmologist with experience in NF2.

Neuroimaging – High-resolution, contrast-enhanced MRI of the brain with thin cuts (<3 mm with no skipping) through the internal auditory canal should be performed for optimal resolution to detect small vestibular schwannomas. Contrast-enhanced MRI of the spine is recommended for all patients at the time of diagnosis to assess the burden of spinal cord involvement and rule out spinal cord impingement.

Diagnostic criteria — The clinical diagnosis of NF2 is based upon the presence of any one of the following criteria [1,51,52]:

Bilateral vestibular schwannomas

An identical pathogenic NF2 variant in at least two anatomically distinct NF2-related tumors (schwannoma, meningioma, and/or ependymoma); if the variant allele fraction in blood is clearly <50 percent, the diagnosis is mosaic NF2

Two of the following major criteria:

Unilateral vestibular schwannoma

First-degree relative other than sibling with NF2

Multiple meningiomas

A pathogenic NF2 variant in nontumorous material (eg, blood)

One major criterion and two of the following minor criteria:

Can be counted twice (eg, two distinct schwannomas would count as two minor criteria):

Nonvestibular schwannoma (at least one dermal if unilateral vestibular schwannoma used as major criterion)

Ependymoma

Can only be counted once:

Juvenile subcapsular or cortical cataract

Retinal hamartoma

Epiretinal membranes

Single meningioma (only if multiple meningiomas not used as major criterion)

Criteria for mosaic NF2 are met in the presence of either of the following:

Clearly <50 percent pathogenic variant allele fraction in blood or saliva

Pathogenic variant not detected in blood or saliva but shared pathogenic variant in two or more anatomically unrelated tumors

Clinical criteria for NF2 were initially established by a National Institutes of Health (NIH) consensus conference [53]. These were subsequently modified into the Manchester criteria, which substantially improved the sensitivity for making the diagnosis without affecting specificity [29,54]. These were further adapted by an international consensus group [1].

Revisions to the NF2 criteria take into account the possibility that unilateral vestibular schwannomas may be due to LZTR1 germline variants (ie, LZTR1-related schwannomatosis) rather than NF2 (see "Schwannomatoses related to genetic variants other than NF2"). Although the diminishing likelihood of NF2 in patients with vestibular schwannomas diagnosed after 70 years of age [51,55] was considered, no age limit was set in the 2022 criteria [1]. In addition, ependymoma has replaced glioma in the diagnostic criteria based on the absence of high-grade gliomas [56] and predominance of ependymomas in NF2 [52].

Genetic testing — Genetic testing is recommended in all patients with suspected schwannomatosis predisposition syndromes but is not required for the diagnosis of NF2 in patients who fulfill clinical criteria. Genetic testing is particularly helpful in younger patients who do not meet diagnostic criteria without genetic data (algorithm 1 and algorithm 2). Genetic testing is also important for first-degree relatives of an individual with NF2, who can be diagnosed with NF2 based on an NF2 pathogenic variant, even in the absence of clinical features.

There is a high frequency of de novo NF2 cases, and patients may present before the clinical diagnostic criteria are fulfilled. Early identification of patients who have NF2 is important to minimize the complications associated with disease manifestations.

Referral to a clinical geneticist or genetic counselor is recommended for patients in whom genetic testing is being considered. Ideally, molecular testing for NF2 should start with tumor deoxyribonucleic acid (DNA) and then targeted testing of blood lymphocytes by sequencing of any NF2 aberration identified. If two tumors are available, identification of an identical NF2 variant in two tumors that is not present in blood confirms mosaicism. LZTR1 should be assessed if individuals meet NF2 criteria with only nonintradermal schwannoma disease. (See 'Diagnostic criteria' above.)

Comprehensive molecular genetic evaluation for NF2 is recommended in all patients suspected of having NF2, including individuals with any of the following [57,58]:

A first-degree relative with NF2 (ie, affected parent, sibling, or offspring)

Multiple spinal tumors (schwannomas, meningiomas)

Cutaneous schwannomas

An apparently sporadic vestibular schwannoma in an individual younger than 30 years of age

A solitary meningioma or nonvestibular schwannoma in an individual younger than 25 years of age [59]

Two or more anatomically distinct meningiomas in an individual aged 25 to 69 years and three or more meningiomas at any age [57,60]

The role of genetic testing for NF2 in all patients with a unilateral vestibular schwannoma is less certain. Although these patients appear to be at increased risk for the development of NF2, routine screening for germline variants does not appear to be indicated except in those younger than 30 years of age [57,58]. There is also evidence that NF2 is unlikely in first-degree relatives who only manifest unilateral vestibular schwannoma [61].

Of note, some individuals have been identified who carry a likely pathogenic variant in NF2 in blood but do not have clinical features of NF2 (eg, in an asymptomatic individual who underwent genetic sequencing for another indication). Given the complete penetrance of NF2 pathogenic variants, such individuals and their at-risk relatives should be referred to a tertiary care center with expertise in schwannomatosis for further evaluation; testing to determine whether the variant is constitutional (germline), mosaic, or somatic; and genetic counseling [1].

Differential diagnosis — The major entities in the differential diagnosis of NF2 are sporadic vestibular schwannomas, other schwannomatoses, neurofibromatosis type 1 (NF1), and a familial syndrome of multiple meningiomas [3].

Sporadic vestibular schwannomas — Sporadic vestibular schwannomas are relatively common in the general population. Although these tumors are unilateral, their diagnosis can cause confusion in some cases. (See "Vestibular schwannoma (acoustic neuroma)".)

The possibility that a unilateral vestibular schwannoma represents the first manifestation of NF2 is related to the age of the patient. For younger individuals who are less than 30 years of age, there is a substantial risk of developing a contralateral vestibular schwannoma, and patients should be monitored carefully for that possibility. Even with a negative family history, these may represent a de novo somatic variant with mosaicism [58,62]. The likelihood that an individual diagnosed with a vestibular schwannoma after the age of 30 will have NF2 is minimal.

In patients with a unilateral vestibular schwannoma and multiple noncutaneous schwannomas, schwannomatosis due to variants in LZTR1 must also be considered. (See 'Other schwannomatoses' below.)

Other schwannomatoses — Distinct from NF2, there are rare sporadic or familial schwannomatoses characterized by multiple noncutaneous peripheral and intracranial schwannomas in the absence of bilateral vestibular schwannomas.

These syndromes are caused by variants in genes other than NF2 on chromosome 22, including SMARCB1 and LZTR1. Among these, LZTR1-related schwannomatosis has the greatest diagnostic overlap with NF2, because individuals may have unilateral vestibular schwannomas, in addition to other tumors [63].

The schwannomas seen in individuals with non-NF2-related schwannomatosis contain somatic variants in the NF2 gene, but such variants are not present in the germline.

The clinical features, diagnosis, and management of non-NF2-related schwannomatoses are reviewed separately. (See "Schwannomatoses related to genetic variants other than NF2".)

Neurofibromatosis type 1 — NF2 and NF1 are caused by variants in genes on different chromosomes. Nonetheless, partial overlap in the clinical manifestations of these inherited disorders can occasionally lead to confusion. (See "Neurofibromatosis type 1 (NF1): Pathogenesis, clinical features, and diagnosis".)

Key differences between NF1 and NF2 include:

Lisch nodules (raised, pigmented hamartomas of the iris) are characteristic of NF1 and are not seen in significant numbers in NF2.

The schwannomas associated with NF2 rarely, if ever, undergo malignant transformation into a neurofibrosarcoma (malignant peripheral nerve sheath tumor [MPNST]) [64].

The "dumbbell" spinal root tumors that are seen with both NF2 and NF1 are schwannomas in NF2 and neurofibromas in NF1.

NF2 is not associated with the cognitive impairment that is often seen with NF1.

Familial meningioma — Familial meningioma is a rare disorder with only a few families described in the literature. This entity is not associated with abnormalities of the NF2 gene.

Multiple meningiomas can occur sporadically, especially in older adults. The occurrence of a single meningioma in a patient less than 25 years of age should prompt evaluation for NF2 [42]. Individuals aged 25 to 69 with at least two anatomically distinct meningiomas and at any age with three meningiomas should also be assessed for NF2 [57,60]. (See 'Genetic testing' above.)

MANAGEMENT — The management of patients with NF2 is complex and involves multiple disciplines to prevent or treat the various complications that may develop. Whenever possible, patients should be cared for by a multidisciplinary team with expertise in the care of NF2-affected individuals and families [65]. Multidisciplinary teams should include, at a minimum, a neurosurgeon, neuro-otologist, neuroradiologist, neurologist or neuro-oncologist, audiologist, specialist nurse, and geneticist.

Tumor surveillance and follow-up — For individuals known to harbor an NF2 pathogenic variant, follow-up should include [2,3,65]:

Annual history and physical examination, including:

Audiology with measurement of pure-tone thresholds and word recognition scores

Ophthalmologic evaluation

Cutaneous examination

Annual brain MRI, beginning at 10 years of age. If baseline imaging shows no characteristic sites of involvement, frequency can be reduced to every two years. If tumors are detected, brain MRI should be performed twice yearly in the first year, then annually. Brain MRI protocol should include high-resolution (1 to 3 mm slice thickness) postcontrast imaging through the internal auditory meatus, preferably in at least two orthogonal planes.

Surveillance spinal MRI every 24 to 36 months beginning at 10 years of age. The interval between scans may be reduced to every three to five years if there is no disease detected on baseline imaging. If tumors are detected, imaging should be repeated in six months to evaluate tumor growth rate.

Whole-body MRI, including brain and spine, is used at some centers for surveillance.

18F-fluorodeoxyglucose positron emission tomography (FDG-PET) is not recommended for routine screening, as the risk of malignant peripheral nerve sheath tumor (MPNST) is extremely small in an unirradiated patient with NF2. In addition, FDG-PET does not discriminate well between a cellular schwannoma and MPNST.

Treatment of vestibular schwannomas — The goal of treatment for vestibular schwannomas in individuals with NF2 is preservation of function and maintenance of quality of life [2]. As such, the identification of a tumor per se is not an indication for treatment, and the potential benefits must be balanced against the risks of active intervention. Treatment is generally indicated when there is a risk of brainstem compression, deterioration of hearing, and/or facial nerve dysfunction [3].

Surgical management — Vestibular schwannomas are generally managed surgically if treatment is indicated, although first-line bevacizumab plays a role in select cases (see 'Bevacizumab' below). Surgery in patients with NF2 can be more complex than in patients with sporadic tumors, since NF2 tumors are often multifocal [35,66]. Vestibular schwannomas may extend to involve fibers of the facial nerve, and there is a significant risk of damage to the facial nerve during surgery [2]. This is particularly problematic, since the facial nerve controls the blink reflex and innervates the lacrimal gland. Loss of the blink reflex or loss of tearing may complicate other eye problems. (See 'Ophthalmic manifestations' above.)

The role of radiation therapy in managing vestibular schwannomas in patients with NF2 is less clear [67-70]. Reports using radiation in patients with NF2 have shown variable outcomes, and long-term follow-up is lacking. Furthermore, surgical resection may be more difficult following stereotactic radiosurgery. There are also concerns that such patients may have an increased risk of second malignancies [71-73]. The risk of second malignancies may also be increased with fractionated radiation therapy, with one report of a rhabdomyosarcoma occurring in the radiation field in an NF2 patient [74].

Bevacizumab — Bevacizumab, a monoclonal antibody against vascular endothelial growth factor (VEGF), can induce both tumor shrinkage and hearing improvement in patients with NF2-associated vestibular schwannomas, and there is growing experience at multidisciplinary NF2 centers with using bevacizumab as a first-line medical therapy for rapidly growing vestibular schwannomas that are a threat to function, before committing a patient to surgery [75].

The efficacy of bevacizumab for progressive vestibular schwannomas in NF2 is supported by retrospective case series and a limited number of prospective single-arm studies [75-85]. In a meta-analysis of eight observational studies in a total of 161 patients with NF2-associated vestibular schwannomas, the best response to bevacizumab was partial regression in 41 percent, no change in 47 percent, and progression in 7 percent [83]. The median treatment duration was 16 months. In the subset of patients with audiometric data, hearing was improved in 20 percent, stable in 69 percent, and worse in 6 percent. The pooled rate of serious toxicity was 17 percent, including one fatal intracranial hemorrhage. The most common toxicities of any severity were amenorrhea (70 percent), proteinuria (43 percent), and hypertension (33 percent).

There may also be a role for bevacizumab in cystic progressive ependymoma, as shown by a systematic review of treatment reports [86].

The dose and schedule of bevacizumab in NF2 has not been standardized. A proposed regimen is bevacizumab 5 to 7.5 mg/kg every two to three weeks for at least six months or until no further tumor response on successive three-month interval MRIs, followed by maintenance therapy at 2.5 to 5 mg/kg every four weeks [75]. A multicenter trial of an alternative, higher-dose "induction" regimen (10 mg/kg every two weeks for six months) offered no clear advantage over the historical experience with lower-dose regimens [84], and higher doses may increase the risk of renal impairment.

Other targeted therapies — The NF2 gene product appears to affect multiple molecular pathways involved in cell growth. An understanding of this provides a potential opportunity for targeted therapy in the treatment of NF2-related tumors.

There are mixed reports on the efficacy of everolimus, an oral inhibitor of the mammalian target of rapamycin complex 1 (mTORC1), for progressive vestibular schwannomas in patients with NF2 [87,88]. Lapatinib, a dual human epidermal growth factor receptor 2 (HER2) and epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, showed evidence of objective activity in 4 out of 17 patients with NF2-related progressive vestibular schwannoma in a small phase II trial [89], whereas erlotinib, an EGFR inhibitor, was not effective in a retrospective series that included 11 patients with NF2 [90]. A phase II trial of the anaplastic lymphoma kinase (ALK) inhibitor brigatinib is underway in the United States [91]. A histone deacetylase inhibitor is also under investigation for treatment of NF2 patients [92].

Hearing impairment — Hearing impairment in patients with NF2 is associated with decreased quality of life and increased risk for social avoidance and isolation, unemployment, and decreased social support [93]. For patients with severe hearing impairment, strategies such as cochlear or brainstem implants may offer some benefit [94]. Pilot studies have demonstrated the feasibility of novel psychosocial interventions delivered to deaf individuals with NF2 via teleconferencing with captioning technology [95]. (See "Hearing amplification in adults", section on 'Cochlear implants'.)

Meningioma management — Meningiomas can arise both at intracranial sites and at intradural, extramedullary sites in the spinal cord. (See "Epidemiology, pathology, clinical features, and diagnosis of meningioma", section on 'Clinical presentation'.)

Many meningiomas in NF2 grow to a certain size and stop, and do not require active treatment. Risk factors for more rapid growth (eg, ≥2 cm3 per year) are similar to those for sporadic meningiomas and include large tumor size, peritumoral edema, absence of calcifications, and isointense or hyperintense T2-weighted MRI signal [96].

For rapidly growing meningiomas and those threatening functional loss, meningiomas are managed surgically when possible. Radiation therapy has been used in patients whose tumors are not surgically accessible or in those in whom only a partial resection is possible [97]. However, long-term follow-up is lacking in this population. As with the use of radiation in patients with vestibular schwannomas, there is concern about the development of secondary neoplasia. (See "Spinal cord tumors", section on 'Meningioma'.)

Targeted therapies are under investigation. Lapatinib showed some activity in a small series of patients with NF2-associated progressive meningiomas [98]. There is little evidence that bevacizumab has activity in NF2-related meningiomas [99].

Intramedullary spinal tumors — Approximately 75 percent of intramedullary spinal tumors are ependymomas. These are often diagnosed with screening imaging studies and usually grow very slowly without symptoms for a prolonged period. Careful neurologic surveillance for evidence of progression is indicated in this situation. If symptoms require intervention, which is relatively uncommon, treatment is generally surgical resection rather than radiation therapy [100]. There is some evidence that cystic elements of the tumors respond to bevacizumab [101].

Screening of at-risk family members — For families in which there is a documented index case of NF2, screening of at-risk family members is important so that a diagnosis can be established in affected individuals as early as possible [2,57].

When a specific pathogenic variant has been identified in the index case, this information provides a 100 percent specific marker to test other family members. Prenatal testing is possible in settings where a known variant has been identified [3]. However, variant screening is not 100 percent sensitive in patients with NF2.

For children who have not inherited NF2, no further testing or prospective evaluation is necessary. Tumor surveillance and follow-up for individuals with an NF2 pathogenic variant is reviewed above. (See 'Tumor surveillance and follow-up' above.)

PROGNOSIS AND NATURAL HISTORY — Individuals typically present with symptoms due to the NF2-related tumors around age 20 to 25 years [15,29]. The diagnosis of NF2 in children is often delayed, since the earliest manifestations may be skin tumors and ocular findings [24,102]. Rare patients are diagnosed as late as the eighth decade of life with unusually indolent tumor growth patterns [103].

Despite surveillance for the development of tumors in patients with NF2 and the aggressive management of any abnormalities that are detected, NF2 is associated with substantial morbidity and shortened survival [2].

The vestibular schwannomas associated with NF2 are more difficult to manage than their sporadic counterparts. In NF2 the lesions are often multifocal, and histologically these are less vascular and more lobular [104]. Because of this and the bilaterality of the tumors, the majority of patients eventually become completely deaf.

Poor balance due to eighth nerve involvement, visual problems, and muscle weakness can all contribute to immobility, and many patients require use of a wheelchair or other assistive devices in early adulthood.

There is frequent loss of lower cranial nerve function leading to swallowing and speech problems, which may contribute to shortened life expectancy [105,106].

Overall survival appears to be shortened in patients with NF2 due to complications of their tumors. In studies from the 1970s and 1980s, the actuarial survival after establishing the diagnosis was 15 years, with an average age at death of 36 years [29,107,108]. Earlier diagnosis and advances in treatment may have improved outcomes [109,110].

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topic (see "Patient education: Vestibular schwannoma (acoustic neuroma) (The Basics)")

SUMMARY AND RECOMMENDATIONS

What is NF2?NF2-related schwannomatosis (NF2; formerly known as neurofibromatosis type 2) is a dominantly inherited syndrome that predisposes individuals to bilateral vestibular schwannomas as well as multiple other tumors of the nervous system. (See 'Introduction' above and 'Terminology' above.)

Pathogenesis – NF2 is due to pathogenic variants in the NF2 gene, which produces merlin, a cell membrane-related protein that acts as a tumor suppressor. (See 'Molecular pathogenesis' above.)

Clinical manifestations – The clinical manifestations of NF2 include bilateral vestibular schwannomas in almost all patients. Other commonly observed nervous system tumors include intracranial meningiomas, schwannomas of other cranial nerves, and spinal tumors (especially schwannomas of the dorsal nerve roots, meningiomas, and ependymomas). Neuropathies and ocular and cutaneous manifestations are also frequent. (See 'Clinical features' above.)

Diagnosis – The diagnosis of NF2 is based upon consensus criteria that incorporate both clinical features and genetic testing. (See 'Clinical and radiologic evaluation' above and 'Diagnostic criteria' above.)

Early identification of patients who have NF2 is important to minimize the complications associated with disease manifestations. Evaluation for NF2, including comprehensive molecular genetic evaluation (algorithm 1 and algorithm 2), is recommended in all patients suspected of having NF2, including individuals with any of the following:

A first-degree relative with NF2 (ie, affected parent, sibling, or offspring)

Multiple spinal tumors (schwannomas, meningiomas)

Cutaneous schwannomas

An apparently sporadic vestibular schwannoma in an individual younger than 30 years of age

A solitary meningioma or nonvestibular schwannoma in an individual younger than 25 years of age

Two or more anatomically distinct meningiomas in an individual aged 25 to 69 years and three or more meningiomas at any age

Differential diagnosis – The differential diagnosis includes sporadic unilateral vestibular schwannomas, other schwannoma predisposition syndromes, neurofibromatosis type 1 (NF1), and familial meningiomas. (See 'Diagnosis' above.)

Management – Whenever possible, patients should be cared for by a multidisciplinary team with expertise in the care of NF2-affected individuals and families. (See 'Management' above.)

All patients with NF2 require brain and spinal tumor surveillance and annual clinical follow-up that includes audiology, ophthalmologic evaluation, neurologic examination, and cutaneous examination. (See 'Tumor surveillance and follow-up' above.)

Vestibular schwannomas are generally managed surgically if treatment is indicated, although first-line bevacizumab plays a role in select cases. For patients with severe hearing impairment, strategies such as cochlear or brainstem implants may offer some benefit. (See 'Treatment of vestibular schwannomas' above and 'Hearing impairment' above.)

Many meningiomas in NF2 grow to a certain size and stop, and do not require active treatment. For rapidly growing meningiomas and those threatening functional loss, meningiomas are managed surgically when possible. (See 'Meningioma management' above.)

Spinal ependymomas are often diagnosed with screening imaging studies and usually grow very slowly without symptoms for a prolonged period. (See 'Intramedullary spinal tumors' above.)

Prognosis – NF2 is associated with substantial morbidity and shortened survival due to complications of central nervous system tumors. (See 'Prognosis and natural history' above.)

  1. Plotkin SR, Messiaen L, Legius E, et al. Updated diagnostic criteria and nomenclature for neurofibromatosis type 2 and schwannomatosis: An international consensus recommendation. Genet Med 2022; 24:1967.
  2. Evans DG. Neurofibromatosis type 2 (NF2): a clinical and molecular review. Orphanet J Rare Dis 2009; 4:16.
  3. Evans DG. Neurofibromatosis 2 [Bilateral acoustic neurofibromatosis, central neurofibromatosis, NF2, neurofibromatosis type II]. Genet Med 2009; 11:599.
  4. Asthagiri AR, Parry DM, Butman JA, et al. Neurofibromatosis type 2. Lancet 2009; 373:1974.
  5. Seizinger BR, Martuza RL, Gusella JF. Loss of genes on chromosome 22 in tumorigenesis of human acoustic neuroma. Nature 1986; 322:644.
  6. Rouleau GA, Merel P, Lutchman M, et al. Alteration in a new gene encoding a putative membrane-organizing protein causes neuro-fibromatosis type 2. Nature 1993; 363:515.
  7. Trofatter JA, MacCollin MM, Rutter JL, et al. A novel moesin-, ezrin-, radixin-like gene is a candidate for the neurofibromatosis 2 tumor suppressor. Cell 1993; 72:791.
  8. Evans DG, Ramsden RT, Shenton A, et al. Mosaicism in neurofibromatosis type 2: an update of risk based on uni/bilaterality of vestibular schwannoma at presentation and sensitive mutation analysis including multiple ligation-dependent probe amplification. J Med Genet 2007; 44:424.
  9. Kluwe L, Mautner V, Heinrich B, et al. Molecular study of frequency of mosaicism in neurofibromatosis 2 patients with bilateral vestibular schwannomas. J Med Genet 2003; 40:109.
  10. Moyhuddin A, Baser ME, Watson C, et al. Somatic mosaicism in neurofibromatosis 2: prevalence and risk of disease transmission to offspring. J Med Genet 2003; 40:459.
  11. Evans DG, Wallace AJ, Wu CL, et al. Somatic mosaicism: a common cause of classic disease in tumor-prone syndromes? Lessons from type 2 neurofibromatosis. Am J Hum Genet 1998; 63:727.
  12. Castellanos E, Plana A, Carrato C, et al. Early Genetic Diagnosis of Neurofibromatosis Type 2 From Skin Plaque Plexiform Schwannomas in Childhood. JAMA Dermatol 2018; 154:341.
  13. Evans DG, Hartley CL, Smith PT, et al. Incidence of mosaicism in 1055 de novo NF2 cases: much higher than previous estimates with high utility of next-generation sequencing. Genet Med 2020; 22:53.
  14. Hadfield KD, Smith MJ, Urquhart JE, et al. Rates of loss of heterozygosity and mitotic recombination in NF2 schwannomas, sporadic vestibular schwannomas and schwannomatosis schwannomas. Oncogene 2010; 29:6216.
  15. Forde C, King AT, Rutherford SA, et al. Disease course of neurofibromatosis type 2: a 30-year follow-up study of 353 patients seen at a single institution. Neuro Oncol 2021; 23:1113.
  16. Selvanathan SK, Shenton A, Ferner R, et al. Further genotype--phenotype correlations in neurofibromatosis 2. Clin Genet 2010; 77:163.
  17. Smith MJ, Higgs JE, Bowers NL, et al. Cranial meningiomas in 411 neurofibromatosis type 2 (NF2) patients with proven gene mutations: clear positional effect of mutations, but absence of female severity effect on age at onset. J Med Genet 2011; 48:261.
  18. Hexter A, Jones A, Joe H, et al. Clinical and molecular predictors of mortality in neurofibromatosis 2: a UK national analysis of 1192 patients. J Med Genet 2015; 52:699.
  19. Halliday D, Emmanouil B, Pretorius P, et al. Genetic Severity Score predicts clinical phenotype in NF2. J Med Genet 2017; 54:657.
  20. Evans DG, Moran A, King A, et al. Incidence of vestibular schwannoma and neurofibromatosis 2 in the North West of England over a 10-year period: higher incidence than previously thought. Otol Neurotol 2005; 26:93.
  21. Evans DG, Howard E, Giblin C, et al. Birth incidence and prevalence of tumor-prone syndromes: estimates from a UK family genetic register service. Am J Med Genet A 2010; 152A:327.
  22. Antinheimo J, Sankila R, Carpén O, et al. Population-based analysis of sporadic and type 2 neurofibromatosis-associated meningiomas and schwannomas. Neurology 2000; 54:71.
  23. Evans DG, Bowers NL, Tobi S, et al. Schwannomatosis: a genetic and epidemiological study. J Neurol Neurosurg Psychiatry 2018; 89:1215.
  24. Evans DG, Birch JM, Ramsden RT. Paediatric presentation of type 2 neurofibromatosis. Arch Dis Child 1999; 81:496.
  25. Anand G, Vasallo G, Spanou M, et al. Diagnosis of sporadic neurofibromatosis type 2 in the paediatric population. Arch Dis Child 2018; 103:463.
  26. Gaudioso C, Listernick R, Fisher MJ, et al. Neurofibromatosis 2 in children presenting during the first decade of life. Neurology 2019; 93:e964.
  27. Parry DM, Eldridge R, Kaiser-Kupfer MI, et al. Neurofibromatosis 2 (NF2): clinical characteristics of 63 affected individuals and clinical evidence for heterogeneity. Am J Med Genet 1994; 52:450.
  28. Mautner VF, Lindenau M, Baser ME, et al. The neuroimaging and clinical spectrum of neurofibromatosis 2. Neurosurgery 1996; 38:880.
  29. Evans DG, Huson SM, Donnai D, et al. A clinical study of type 2 neurofibromatosis. Q J Med 1992; 84:603.
  30. Bosch MM, Boltshauser E, Harpes P, Landau K. Ophthalmologic findings and long-term course in patients with neurofibromatosis type 2. Am J Ophthalmol 2006; 141:1068.
  31. Ragge NK, Baser ME, Riccardi VM, Falk RE. The ocular presentation of neurofibromatosis 2. Eye (Lond) 1997; 11 ( Pt 1):12.
  32. Ragge NK, Baser ME, Klein J, et al. Ocular abnormalities in neurofibromatosis 2. Am J Ophthalmol 1995; 120:634.
  33. MacCollin M, Mautner VF. The diagnosis and management of neurofibromatosis 2 in childhood. Semin Pediatr Neurol 1998; 5:243.
  34. Mautner VF, Lindenau M, Baser ME, et al. Skin abnormalities in neurofibromatosis 2. Arch Dermatol 1997; 133:1539.
  35. Evans DG, Stivaros SM. Multifocality in neurofibromatosis type 2. Neuro Oncol 2015; 17:481.
  36. Abaza MM, Makariou E, Armstrong M, Lalwani AK. Growth rate characteristics of acoustic neuromas associated with neurofibromatosis type 2. Laryngoscope 1996; 106:694.
  37. Fisher LM, Doherty JK, Lev MH, Slattery WH. Concordance of bilateral vestibular schwannoma growth and hearing changes in neurofibromatosis 2: neurofibromatosis 2 natural history consortium. Otol Neurotol 2009; 30:835.
  38. Masuda A, Fisher LM, Oppenheimer ML, et al. Hearing changes after diagnosis in neurofibromatosis type 2. Otol Neurotol 2004; 25:150.
  39. Graamans K, Van Dijk JE, Janssen LW. Hearing deterioration in patients with a non-growing vestibular schwannoma. Acta Otolaryngol 2003; 123:51.
  40. Asthagiri AR, Vasquez RA, Butman JA, et al. Mechanisms of hearing loss in neurofibromatosis type 2. PLoS One 2012; 7:e46132.
  41. Goutagny S, Kalamarides M. Meningiomas and neurofibromatosis. J Neurooncol 2010; 99:341.
  42. Evans DG, Watson C, King A, et al. Multiple meningiomas: differential involvement of the NF2 gene in children and adults. J Med Genet 2005; 42:45.
  43. Perry A, Giannini C, Raghavan R, et al. Aggressive phenotypic and genotypic features in pediatric and NF2-associated meningiomas: a clinicopathologic study of 53 cases. J Neuropathol Exp Neurol 2001; 60:994.
  44. Larson JJ, van Loveren HR, Balko MG, Tew JM Jr. Evidence of meningioma infiltration into cranial nerves: clinical implications for cavernous sinus meningiomas. J Neurosurg 1995; 83:596.
  45. Patronas NJ, Courcoutsakis N, Bromley CM, et al. Intramedullary and spinal canal tumors in patients with neurofibromatosis 2: MR imaging findings and correlation with genotype. Radiology 2001; 218:434.
  46. Dow G, Biggs N, Evans G, et al. Spinal tumors in neurofibromatosis type 2. Is emerging knowledge of genotype predictive of natural history? J Neurosurg Spine 2005; 2:574.
  47. Godel T, Bäumer P, Farschtschi S, et al. Peripheral nervous system alterations in infant and adult neurofibromatosis type 2. Neurology 2019; 93:e590.
  48. Sperfeld AD, Hein C, Schröder JM, et al. Occurrence and characterization of peripheral nerve involvement in neurofibromatosis type 2. Brain 2002; 125:996.
  49. McLaughlin ME, Pepin SM, Maccollin M, et al. Ocular pathologic findings of neurofibromatosis type 2. Arch Ophthalmol 2007; 125:389.
  50. Painter SL, Sipkova Z, Emmanouil B, et al. Neurofibromatosis Type 2-Related Eye Disease Correlated With Genetic Severity Type. J Neuroophthalmol 2019; 39:44.
  51. Smith MJ, Bowers NL, Bulman M, et al. Revisiting neurofibromatosis type 2 diagnostic criteria to exclude LZTR1-related schwannomatosis. Neurology 2017; 88:87.
  52. Evans DG, King AT, Bowers NL, et al. Identifying the deficiencies of current diagnostic criteria for neurofibromatosis 2 using databases of 2777 individuals with molecular testing. Genet Med 2019; 21:1525.
  53. Neurofibromatosis. Conference statement. National Institutes of Health Consensus Development Conference. Arch Neurol 1988; 45:575.
  54. Gutmann DH, Aylsworth A, Carey JC, et al. The diagnostic evaluation and multidisciplinary management of neurofibromatosis 1 and neurofibromatosis 2. JAMA 1997; 278:51.
  55. Evans DG, Freeman S, Gokhale C, et al. Bilateral vestibular schwannomas in older patients: NF2 or chance? J Med Genet 2015; 52:422.
  56. King AT, Rutherford SA, Hammerbeck-Ward C, et al. High-Grade Glioma is not a Feature of Neurofibromatosis Type 2 in the Unirradiated Patient. Neurosurgery 2018; 83:193.
  57. Evans DG, Raymond FL, Barwell JG, Halliday D. Genetic testing and screening of individuals at risk of NF2. Clin Genet 2012; 82:416.
  58. Evans DG, Ramsden RT, Gokhale C, et al. Should NF2 mutation screening be undertaken in patients with an apparently isolated vestibular schwannoma? Clin Genet 2007; 71:354.
  59. Pathmanaban ON, Sadler KV, Kamaly-Asl ID, et al. Association of Genetic Predisposition With Solitary Schwannoma or Meningioma in Children and Young Adults. JAMA Neurol 2017; 74:1123.
  60. Hannan CJ, Hammerbeck-Ward C, Pathmanaban ON, et al. Multiple Meningiomas as a Criterion for the Diagnosis of Neurofibromatosis Type 2 and Other Tumor Predisposition Syndromes. Neurosurgery 2022; 90:793.
  61. Evans DG, Wallace AJ, Hartley C, et al. Familial unilateral vestibular schwannoma is rarely caused by inherited variants in the NF2 gene. Laryngoscope 2019; 129:967.
  62. Mohyuddin A, Neary WJ, Wallace A, et al. Molecular genetic analysis of the NF2 gene in young patients with unilateral vestibular schwannomas. J Med Genet 2002; 39:315.
  63. Smith MJ, Isidor B, Beetz C, et al. Mutations in LZTR1 add to the complex heterogeneity of schwannomatosis. Neurology 2015; 84:141.
  64. King AT, Rutherford SA, Hammerbeck-Ward C, et al. Malignant Peripheral Nerve Sheath Tumors are not a Feature of Neurofibromatosis Type 2 in the Unirradiated Patient. Neurosurgery 2018; 83:38.
  65. Evans DGR, Salvador H, Chang VY, et al. Cancer and Central Nervous System Tumor Surveillance in Pediatric Neurofibromatosis 2 and Related Disorders. Clin Cancer Res 2017; 23:e54.
  66. Dewan R, Pemov A, Kim HJ, et al. Evidence of polyclonality in neurofibromatosis type 2-associated multilobulated vestibular schwannomas. Neuro Oncol 2015; 17:566.
  67. Kida Y, Kobayashi T, Tanaka T, Mori Y. Radiosurgery for bilateral neurinomas associated with neurofibromatosis type 2. Surg Neurol 2000; 53:383.
  68. Mathieu D, Kondziolka D, Flickinger JC, et al. Stereotactic radiosurgery for vestibular schwannomas in patients with neurofibromatosis type 2: an analysis of tumor control, complications, and hearing preservation rates. Neurosurgery 2007; 60:460.
  69. Roche PH, Régis J, Pellet W, et al. [Neurofibromatosis type 2. Preliminary results of gamma knife radiosurgery of vestibular schwannomas]. Neurochirurgie 2000; 46:339.
  70. Rowe JG, Radatz MW, Walton L, et al. Clinical experience with gamma knife stereotactic radiosurgery in the management of vestibular schwannomas secondary to type 2 neurofibromatosis. J Neurol Neurosurg Psychiatry 2003; 74:1288.
  71. Baser ME, Evans DG, Jackler RK, et al. Neurofibromatosis 2, radiosurgery and malignant nervous system tumours. Br J Cancer 2000; 82:998.
  72. Evans DG, Birch JM, Ramsden RT, et al. Malignant transformation and new primary tumours after therapeutic radiation for benign disease: substantial risks in certain tumour prone syndromes. J Med Genet 2006; 43:289.
  73. Balasubramaniam A, Shannon P, Hodaie M, et al. Glioblastoma multiforme after stereotactic radiotherapy for acoustic neuroma: case report and review of the literature. Neuro Oncol 2007; 9:447.
  74. Carlson ML, Babovic-Vuksanovic D, Messiaen L, et al. Radiation-induced rhabdomyosarcoma of the brainstem in a patient with neurofibromatosis type 2. J Neurosurg 2010; 112:81.
  75. Morris KA, Golding JF, Axon PR, et al. Bevacizumab in neurofibromatosis type 2 (NF2) related vestibular schwannomas: a nationally coordinated approach to delivery and prospective evaluation. Neurooncol Pract 2016; 3:281.
  76. Mautner VF, Nguyen R, Kutta H, et al. Bevacizumab induces regression of vestibular schwannomas in patients with neurofibromatosis type 2. Neuro Oncol 2010; 12:14.
  77. Plotkin SR, Stemmer-Rachamimov AO, Barker FG 2nd, et al. Hearing improvement after bevacizumab in patients with neurofibromatosis type 2. N Engl J Med 2009; 361:358.
  78. Plotkin SR, Merker VL, Halpin C, et al. Bevacizumab for progressive vestibular schwannoma in neurofibromatosis type 2: a retrospective review of 31 patients. Otol Neurotol 2012; 33:1046.
  79. Hochart A, Gaillard V, Baroncini M, et al. Bevacizumab decreases vestibular schwannomas growth rate in children and teenagers with neurofibromatosis type 2. J Neurooncol 2015; 124:229.
  80. Blakeley JO, Ye X, Duda DG, et al. Efficacy and Biomarker Study of Bevacizumab for Hearing Loss Resulting From Neurofibromatosis Type 2-Associated Vestibular Schwannomas. J Clin Oncol 2016; 34:1669.
  81. Sverak P, Adams ME, Haines SJ, et al. Bevacizumab for Hearing Preservation in Neurofibromatosis Type 2: Emphasis on Patient-Reported Outcomes and Toxicities. Otolaryngol Head Neck Surg 2019; 160:526.
  82. Li KL, Djoukhadar I, Zhu X, et al. Vascular biomarkers derived from dynamic contrast-enhanced MRI predict response of vestibular schwannoma to antiangiogenic therapy in type 2 neurofibromatosis. Neuro Oncol 2016; 18:275.
  83. Lu VM, Ravindran K, Graffeo CS, et al. Efficacy and safety of bevacizumab for vestibular schwannoma in neurofibromatosis type 2: a systematic review and meta-analysis of treatment outcomes. J Neurooncol 2019; 144:239.
  84. Plotkin SR, Duda DG, Muzikansky A, et al. Multicenter, Prospective, Phase II and Biomarker Study of High-Dose Bevacizumab as Induction Therapy in Patients With Neurofibromatosis Type 2 and Progressive Vestibular Schwannoma. J Clin Oncol 2019; 37:3446.
  85. Plotkin SR, Allen J, Dhall G, et al. Multicenter, prospective, phase II study of maintenance bevacizumab for children and adults with NF2-related schwannomatosis and progressive vestibular schwannoma. Neuro Oncol 2023; 25:1498.
  86. Snyder MH, Ampie L, DiDomenico JD, Asthagiri AR. Bevacizumab as a surgery-sparing agent for spinal ependymoma in patients with neurofibromatosis type II: Systematic review and case. J Clin Neurosci 2021; 86:79.
  87. Goutagny S, Raymond E, Esposito-Farese M, et al. Phase II study of mTORC1 inhibition by everolimus in neurofibromatosis type 2 patients with growing vestibular schwannomas. J Neurooncol 2015; 122:313.
  88. Karajannis MA, Legault G, Hagiwara M, et al. Phase II study of everolimus in children and adults with neurofibromatosis type 2 and progressive vestibular schwannomas. Neuro Oncol 2014; 16:292.
  89. Karajannis MA, Legault G, Hagiwara M, et al. Phase II trial of lapatinib in adult and pediatric patients with neurofibromatosis type 2 and progressive vestibular schwannomas. Neuro Oncol 2012; 14:1163.
  90. Plotkin SR, Halpin C, McKenna MJ, et al. Erlotinib for progressive vestibular schwannoma in neurofibromatosis 2 patients. Otol Neurotol 2010; 31:1135.
  91. Innovative Trial for Understanding the Impact of Targeted Therapies in NF2 (INTUITT-NF2). ClinicalTrials.gov. Available at: https://clinicaltrials.gov/ct2/show/NCT04374305 (Accessed on May 12, 2021).
  92. Collier KA, Valencia H, Newton H, et al. A phase 1 trial of the histone deacetylase inhibitor AR-42 in patients with neurofibromatosis type 2-associated tumors and advanced solid malignancies. Cancer Chemother Pharmacol 2021; 87:599.
  93. Merker VL, Bergner AL, Vranceanu AM, et al. Health-related Quality of Life of Individuals With Neurofibromatosis Type 2: Results From the NF2 Natural History Study. Otol Neurotol 2016; 37:574.
  94. Neff BA, Wiet RM, Lasak JM, et al. Cochlear implantation in the neurofibromatosis type 2 patient: long-term follow-up. Laryngoscope 2007; 117:1069.
  95. Funes CJ, Mace RA, Macklin EA, et al. First report of quality of life in adults with neurofibromatosis 2 who are deafened or have significant hearing loss: results of a live-video randomized control trial. J Neurooncol 2019; 143:505.
  96. Abi Jaoude S, Peyre M, Degos V, et al. Validation of a scoring system to evaluate the risk of rapid growth of intracranial meningiomas in neurofibromatosis type 2 patients. J Neurosurg 2020; 134:1377.
  97. Wentworth S, Pinn M, Bourland JD, et al. Clinical experience with radiation therapy in the management of neurofibromatosis-associated central nervous system tumors. Int J Radiat Oncol Biol Phys 2009; 73:208.
  98. Osorio DS, Hu J, Mitchell C, et al. Effect of lapatinib on meningioma growth in adults with neurofibromatosis type 2. J Neurooncol 2018; 139:749.
  99. Nunes FP, Merker VL, Jennings D, et al. Bevacizumab treatment for meningiomas in NF2: a retrospective analysis of 15 patients. PLoS One 2013; 8:e59941.
  100. Kalamarides M, Essayed W, Lejeune JP, et al. Spinal ependymomas in NF2: a surgical disease? J Neurooncol 2018; 136:605.
  101. Farschtschi S, Merker VL, Wolf D, et al. Bevacizumab treatment for symptomatic spinal ependymomas in neurofibromatosis type 2. Acta Neurol Scand 2016; 133:475.
  102. Ruggieri M, Iannetti P, Polizzi A, et al. Earliest clinical manifestations and natural history of neurofibromatosis type 2 (NF2) in childhood: a study of 24 patients. Neuropediatrics 2005; 36:21.
  103. Goutagny S, Bah AB, Parfait B, et al. Neurofibromatosis type 2 in the elderly population: clinical and molecular features. Am J Med Genet A 2013; 161A:667.
  104. Sobel RA. Vestibular (acoustic) schwannomas: histologic features in neurofibromatosis 2 and in unilateral cases. J Neuropathol Exp Neurol 1993; 52:106.
  105. Rajendran S, Antonios J, Solomon B, et al. A Prospective Evaluation of Swallowing and Speech in Patients with Neurofibromatosis Type 2. J Neurol Surg B Skull Base 2021; 82:244.
  106. Fisher LM, Doherty JK, Lev MH, Slattery WH 3rd. Distribution of nonvestibular cranial nerve schwannomas in neurofibromatosis 2. Otol Neurotol 2007; 28:1083.
  107. Evans DG, Huson SM, Donnai D, et al. A genetic study of type 2 neurofibromatosis in the United Kingdom. I. Prevalence, mutation rate, fitness, and confirmation of maternal transmission effect on severity. J Med Genet 1992; 29:841.
  108. Evans DG, Huson SM, Donnai D, et al. A genetic study of type 2 neurofibromatosis in the United Kingdom. II. Guidelines for genetic counselling. J Med Genet 1992; 29:847.
  109. Evans DG, Baser ME, O'Reilly B, et al. Management of the patient and family with neurofibromatosis 2: a consensus conference statement. Br J Neurosurg 2005; 19:5.
  110. Baser ME, Friedman JM, Aeschliman D, et al. Predictors of the risk of mortality in neurofibromatosis 2. Am J Hum Genet 2002; 71:715.
Topic 5206 Version 34.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟