Von Hippel-Lindau Syndrome

Synonyms: VHL Disease, VHL Syndrome, Von Hippel-Lindau Disease

van Leeuwaarde RS, Ahmad S, van Nesselrooij B, et al.

Publication Details

Estimated reading time: 53 minutes

Summary

Clinical characteristics.

Von Hippel-Lindau syndrome (VHL) is characterized by hemangioblastomas of the brain, spinal cord, and retina; renal cysts and clear cell renal cell carcinoma; pheochromocytoma and paraganglioma; pancreatic cysts and neuroendocrine tumors; endolymphatic sac tumors; and epididymal and broad ligament cystadenomas. Retinal hemangioblastomas may be the initial manifestation of VHL and can cause vision loss. Cerebellar hemangioblastomas may be associated with headache, vomiting, gait disturbances, or ataxia. Spinal hemangioblastomas and related syrinx usually present with pain. Sensory and motor loss may develop with cord compression. Renal cell carcinoma occurs in about 70% of individuals with VHL and is the leading cause of mortality. Pheochromocytomas can be asymptomatic but may cause sustained or episodic hypertension. Pancreatic lesions often remain asymptomatic and rarely cause endocrine or exocrine insufficiency. Endolymphatic sac tumors can cause hearing loss of varying severity, which can be a presenting symptom. Cystadenomas of the epididymis are relatively common. They rarely cause problems, unless bilateral, in which case they may result in infertility.

Diagnosis/testing.

The diagnosis of VHL is established in a proband who fulfills existing diagnostic clinical criteria. Identification of a heterozygous germline VHL pathogenic variant on molecular genetic testing establishes the diagnosis if clinical features are inconclusive.

Management.

Targeted therapies: Pazopanib is an FDA-approved treatment for advanced renal cell carcinoma. Belzutifan is approved in many countries for the treatment of adults with VHL who do not require immediate surgery for associated renal cell carcinoma, central nervous system (CNS) hemangioblastomas, or pancreatic neuroendocrine tumors.

Supportive care: Surgical resection for most CNS hemangioblastomas; early treatment for retinal hemangioblastomas; cryoablation or radiofrequency ablation for renal cell carcinoma; kidney transplantation following bilateral nephrectomy; removal of pheochromocytomas (partial adrenalectomy when possible); consider removal of pancreatic neuroendocrine tumors; consider surgical removal of endolymphatic sac tumors (particularly small tumors in order to preserve hearing and vestibular function); cystadenomas of the epididymis or broad ligament need treatment when symptomatic or threatening fertility; psychosocial support and care coordination as needed.

Surveillance: For individuals with VHL and at-risk relatives of unknown genetic status: annual clinical evaluation for neurologic symptoms, vision problems, and hearing disturbances beginning in the first decade of life; brain and total spine MRI every two years starting at age 11 years; ophthalmology evaluation beginning at age one year; abdominal MRI every two years starting at age 15 years; annual blood pressure starting in the first decade of life; annual plasma or 24-hour urine for fractionated metanephrines starting at age five years; audiology assessment every two to three years starting at age 11 years; MRI of the internal auditory canal in asymptomatic individuals between age 15 and 20 years; assessment of psychosocial needs at each visit.

Agents/circumstances to avoid: Tobacco products should be avoided, as they are considered a risk factor for kidney cancer; chemicals and industrial toxins known to affect VHL-involved organs should be avoided; contact sports should be avoided if adrenal or pancreatic lesions are present.

Evaluation of relatives at risk: If the pathogenic variant in a family is known, molecular genetic testing can be used to clarify the genetic status of at-risk family members to eliminate the need for surveillance of family members who have not inherited the pathogenic variant.

Pregnancy management: Intensified surveillance for cerebellar hemangioblastoma and pheochromocytoma prior to conception and during pregnancy; MRI without contrast of the cerebellum at four months' gestation.

Genetic counseling.

VHL is inherited in an autosomal dominant manner. Approximately 80% of individuals with VHL have an affected parent and about 20% have VHL as the result of a pathogenic variant that occurred as a de novo event in the affected individual or as a postzygotic de novo event in a mosaic, apparently unaffected parent. The offspring of an individual with VHL are at a 50% risk of inheriting the VHL pathogenic variant. Once the VHL pathogenic variant has been identified in an affected family member, testing of at-risk asymptomatic family members, prenatal testing, and preimplantation genetic testing for VHL are possible.

Diagnosis

Clinical diagnostic criteria for von Hippel-Lindau syndrome (VHL) have been proposed and vary slightly between Dutch or Danish guidelines [Binderup et al 2022, Wolters et al 2022].

Suggestive Findings

VHL should be suspected in individuals with or without a family history of VHL who have:

  • Retinal angioma, especially in a young individual
  • Multiple spinal or cerebellar hemangioblastoma, or a single hemangioblastoma diagnosed at age ≤50 years
  • Adrenal or extra-adrenal pheochromocytoma
  • Renal cell carcinoma diagnosed at age ≤40 years
  • Multiple renal and pancreatic cysts
  • Multiple neuroendocrine tumors of the pancreas
  • Endolymphatic sac tumors
  • Less commonly, multiple papillary cystadenomas of the epididymis or broad ligament

Establishing the Diagnosis

The clinical diagnosis of VHL can be established in proband according to Danish or Dutch guidelines (see Table 1), or the molecular diagnosis can be established by identification of a heterozygous pathogenic (or likely pathogenic) variant in VHL on molecular genetic testing (see Table 2).

Note: (1) Per ACMG/AMP variant interpretation guidelines, the terms "pathogenic variant" and "likely pathogenic variant" are synonymous in a clinical setting, meaning that both are considered diagnostic and can be used for clinical decision making [Richards et al 2015]. Reference to "pathogenic variants" in this GeneReview is understood to include likely pathogenic variants. (2) Identification of a heterozygous VHL variant of uncertain significance does not establish or rule out the diagnosis.

Table 1.

Table 1.

Von Hippel-Lindau Syndrome Clinical Diagnostic Criteria

Molecular genetic testing approaches can include a combination of gene-targeted testing (single gene testing, multigene panel) and comprehensive genomic testing (exome sequencing, genome sequencing). Gene-targeted testing requires that the clinician determine which gene(s) are likely involved (see Option 1), whereas comprehensive genomic testing does not (see Option 2).

Option 1

Single-gene testing. Sequence analysis of the VHL coding region, intron 1, and flanking sequences is performed first to detect missense, nonsense, and splice site variants and small intragenic deletions/insertions. Note: Depending on the sequencing method used, single-exon, multiexon, or whole-gene deletions/duplications may not be detected. If no variant is detected by the sequencing method used, the next step is to perform gene-targeted deletion/duplication analysis to detect exon and whole-gene deletions or duplications.

A multigene panel that includes VHL and other genes of interest (see Differential Diagnosis) may be considered to identify the genetic cause of the condition while limiting identification of variants of uncertain significance and pathogenic variants in genes that do not explain the underlying phenotype. Note: (1) The genes included in the panel and the diagnostic sensitivity of the testing used for each gene vary by laboratory and are likely to change over time. (2) Some multigene panels may include genes not associated with the condition discussed in this GeneReview. (3) In some laboratories, panel options may include a custom laboratory-designed panel and/or custom phenotype-focused exome analysis that includes genes specified by the clinician. (4) Methods used in a panel may include sequence analysis, deletion/duplication analysis, and/or other non-sequencing-based tests.

For an introduction to multigene panels click here. More detailed information for clinicians ordering genetic tests can be found here.

Option 2

Comprehensive genomic testing does not require the clinician to determine which gene is likely involved. Exome sequencing is most commonly used; genome sequencing is also possible.

For an introduction to comprehensive genomic testing click here. More detailed information for clinicians ordering genomic testing can be found here.

Table 2.

Table 2.

Molecular Genetic Testing Used in von Hippel-Lindau Syndrome

Clinical Characteristics

Clinical Description

Von Hippel-Lindau syndrome (VHL) is characterized by hemangioblastomas of the brain, spinal cord, and retina; renal cysts and renal cell carcinoma; pheochromocytoma and paraganglioma; pancreatic cysts and neuroendocrine tumors; endolymphatic sac tumors; and epididymal and broad ligament cysts. Some clustering of tumors occurs, resulting in the designation of specific VHL phenotypes. The manifestations and severity are highly variable both within and between families, even among those with the same pathogenic variant.

Hemangioblastomas. Central nervous system (CNS) hemangioblastoma is the prototypic lesion of VHL [Catapano et al 2005, Gläsker 2005]. Multiple CNS tumors, occurring either synchronously or metachronously, are common. Roughly 80% develop in the brain and 20% in the spinal cord. Peripheral nerve hemangiomas may be a rare manifestation [Giannini et al 1998]. Some hemangioblastomas do not cause symptoms and are discovered only on imaging.

Growth patterns of CNS hemangioblastomas can be saltatory (72%), linear (6%), or exponential (22%). Increased growth was associated with male sex, symptomatic tumors, hemangioblastoma-associated cysts [Takami et al 2022], and germline VHL intragenic deletions [Lonser et al 2014, Huntoon et al 2015]. CNS hemangioblastomas remain the main cause of death, although VHL-related survival has improved over time [Binderup et al 2017b].

  • Brain hemangioblastomas. Within the brain, the vast majority are infratentorial, mainly in the cerebellar hemispheres. The pituitary stalk is the most common site for the development of supratentorial hemangioblastomas in individuals with VHL [Lonser et al 2009]. Clinical symptoms depend on the site of the tumor: with infratentorial tumors, headache, vomiting, and gait disturbances or ataxia predominate; with tumors above the tentorium, symptoms depend on the location of the lesion.
  • Spinal hemangioblastomas are generally intradural, most commonly occur in the cervical or thoracic regions, and occasionally may involve the entire spinal cord. Most symptom-producing spinal hemangioblastomas are associated with cysts/syringomyelia/syrinx [Wanebo et al 2003]. Spinal hemangioblastomas usually present with pain; sensory and motor loss may develop with cord compression.
  • Retinal hemangioblastoma, sometimes called retinal angiomas, are histologically identical to CNS hemangioblastomas. They may be the initial manifestations of VHL and may occur in childhood. About 70% of affected individuals are identified as having retinal hemangioblastomas [Webster et al 1999, Kreusel 2005], with mean age of detection about 25 years [Dollfus et al 2002]. The tumors are most often located in the temporal periphery of the retina with feeder and draining vessels going to and from the optic disc. However, they may develop in the posterior pole (1%) and optic disc (8%).
    Retinal hemangioblastomas may be asymptomatic and may be detected on routine ophthalmoscopy. Others present with a visual field defect or a loss of visual activity resulting from retinal detachment, exudation, or hemorrhage. Tests of retinal function may be abnormal even in the presence of quiescent retinal hemangioblastomas [Kreusel et al 2006]. While the number of retinal hemangioblastomas does not appear to increase with age, the probability of vision loss does [Kreusel et al 2006].

Renal lesions

  • Multiple and bilateral renal cysts are common in individuals with VHL [Lonser et al 2003].
  • Renal cell carcinoma, specifically of the clear cell subtype, developing either within a cyst or in the surrounding parenchyma, occurs in about 70% of affected individuals by age 60 years, and is a leading cause of mortality in VHL [Maher et al 1990, Maher et al 1991]. Pathogenic variants in VHL are the most common cause of renal cell carcinoma. Overall survival for renal cell carcinoma in individuals with VHL is associated with tumor size and age of the individual [Kwon et al 2014]. Missense variants seem to lead to faster growth [Peng et al 2019].

Pheochromocytoma may cause sustained or episodic hypertension or may not cause signs/symptoms. Due to surveillance in individuals with VHL, VHL-related pheochromocytoma are more likely to be identified at a younger age and detected incidentally by abdominal imaging in asymptomatic normotensive individuals [Li et al 2020]. Pheochromocytomas can be unilateral or bilateral and are often small and multifocal [Li et al 2020]. They are usually benign, but malignant behavior has been reported [Chen et al 2001, Jimenez et al 2009].

Paragangliomas. Similar in etiology, paragangliomas can develop along the sympathetic axis in the abdomen or thorax [Schimke et al 1998, Boedeker et al 2014]; these tumors are often nonfunctional (i.e., do not secrete catecholamines or other hormones).

Pancreatic lesions

  • Pancreatic cysts. Most pancreatic lesions are simple cysts and have no malignant potential. While they can be numerous in individuals with VHL, they rarely cause endocrine or exocrine insufficiency. Occasionally, cysts in the head of the pancreas cause biliary obstruction.
  • Neuroendocrine tumors. 5%-17% of individuals with VHL develop neuroendocrine tumors of the pancreas [Lonser et al 2003, Maher et al 2011]. They are not usually hormonally active and are slow growing, but malignant behavior has been observed, particularly in tumors >2.7 cm [Krauss et al 2018]. Tumors have been described from an early age, starting from the second decade [Krauss et al 2018, O'Toole et al 2018].

Endolymphatic sac tumors are seen in approximately 10%-16% of individuals with VHL, and in some instances unilateral or bilateral hearing loss is the initial clinical manifestation of VHL [Kim et al 2005, Binderup et al 2013]. The onset of hearing loss is typically sudden; severity varies, but it is often severe to profound [Choo et al 2004, Kim et al 2005]. Vertigo or tinnitus can be the initial manifestation of an endolymphatic sac tumor. More significant hearing loss and larger tumor size at presentation was reported in individuals with endolymphatic sac tumors not related to VHL than in individuals with VHL-related endolymphatic sac tumors [Nevoux et al 2014]. Large endolymphatic sac tumors can involve other cranial nerves. Endolymphatic sac tumors are rarely malignant [Muzumdar et al 2006].

Epididymal and broad ligament cystadenomas. Epididymal or papillary cystadenomas are relatively common in males with VHL. They rarely cause problems, unless bilateral, in which case they may result in infertility. The equivalent, much less common, lesion in women is a papillary cystadenoma of the broad ligament. Both tissues are mesonephric in origin and are likely a developmental remnant of somatic VHL loss.

Prognosis. One study showed that in adulthood, men have more VHL manifestations compared to women, and the risk for manifestations was not constant but varied throughout the affected individual's lifetime [Binderup et al 2016]. Age was the only predictor for the number of involved organs.

Improved surveillance guidelines have increased the life expectancy of individuals with VHL by more than 16 years since 1990 [Wilding et al 2012]. Two studies evaluated the implementation of national surveillance guidelines in Denmark and the Netherlands. One study of 84 individuals with VHL showed that more than 90% reported that they were familiar with their national VHL surveillance guidelines. However, 64% of those individuals had received information that was only partially consistent with the Dutch guidelines [Lammens et al 2011a]. In a Danish study, compliance and frequency of recommended surveillance was low for individuals with VHL and individuals at risk [Bertelsen & Kosteljanetz 2011]. These studies suggest that implementation of surveillance guidelines through a doctor- and patient-oriented information campaign could have an immediate positive impact for individuals with VHL.

Genotype-Phenotype Correlations

Four general VHL phenotypes (type 1, type 2A, type 2B, type 2C) have been suggested based on the likelihood of pheochromocytoma or renal cell carcinoma. Many lines of research support the conclusion that the molecular etiology of pheochromocytomas appears to be distinct from other VHL lesions. Therefore, the most relevant genotype-phenotype correlations rely mostly on scoring the presence/absence of pheochromocytomas associated with a given allele. The following discussion summarizes the genotype-phenotype studies published to date, with the cautionary note that further investigation is needed. Note: Patterns are not clear-cut, and genotype-phenotype correlations have no current diagnostic or therapeutic value; they are used for academic purposes only.

VHL type 1. Retinal angioma, CNS hemangioblastoma, renal cell carcinoma, pancreatic cysts, and neuroendocrine tumors. VHL type 1 is characterized by a low risk for pheochromocytoma. Pathogenic truncating or missense variants that are predicted to grossly disrupt the folding of the VHL protein [Stebbins et al 1999] are associated with VHL type 1.

Several groups report a reduced risk for renal cell carcinoma in individuals with a deletion of VHL [Cybulski et al 2002, Maranchie et al 2004, McNeill et al 2009]. In particular, individuals with a complete or partial deletion that extends 5' of VHL to include BRK1 (previously C3orf10) have a significantly reduced risk of renal cell carcinoma [Maranchie et al 2004, McNeill et al 2009]. This genotype may constitute a distinct phenotype, VHL type 1B, characterized by a reduced risk for both renal cell carcinoma and pheochromocytoma.

VHL type 2. Pheochromocytoma, retinal hemangioblastomas, and CNS hemangioblastoma. VHL type 2 is characterized by a high risk for pheochromocytoma. Individuals with VHL type 2 commonly have a pathogenic missense variant. Some pathogenic missense variants appear to correlate with a specific VHL type 2 phenotype [Weirich et al 2002, Sansó et al 2004, Abbott et al 2006, Knauth et al 2006] (see also Molecular Genetics). Pathogenic missense variants stratified by multiple in silico computational models found that variants with a high predicted risk of pathogenicity were predictive of pancreatic lesion progression [Tirosh et al 2018a]. In contrast, genotype did not appear to influence the growth of renal cell carcinomas in individuals with VHL [Farhadi et al 2018].

VHL type 2 is further subdivided:

  • Type 2A. Pheochromocytoma, retinal hemangioblastomas, and CNS hemangioblastoma; low risk for renal cell carcinoma
  • Type 2B. Pheochromocytoma, retinal hemangioblastomas, CNS hemangioblastoma, pancreatic cysts, and neuroendocrine tumors; high risk for renal carcinoma
  • Type 2C. Risk for pheochromocytomas only. Note: Some individuals within families with an apparent type 2C phenotype have developed hemangioblastomas [Neumann & Eng 2009].

A higher age-related incidence of retinal hemangioblastomas, CNS hemangioblastomas, clear cell renal cell carcinoma, and pancreatic neuroendocrine tumors was described in individuals with a truncating variant compared to individuals with a missense variant or in-frame deletion of a single amino acid [Reich et al 2021].

Penetrance

VHL pathogenic variants are highly penetrant. Almost all individuals who have a VHL pathogenic variant are symptomatic by age 65 years [Maher et al 1991].

Nomenclature

Obsolete terms for VHL include: angiophakomatosis retinae et cerebelli, familial cerebello-retinal angiomatosis, cerebelloretinal hemangioblastomatosis, Hippel disease, Hippel-Lindau syndrome, Lindau disease, and retinocerebellar angiomatosis [Molino et al 2006].

Prevalence

The incidence of VHL is thought to be about one in 36,000 births, with an estimated de novo mutation rate of 4.4 x 10-6 gametes per generation [Maher et al 1991].

Differential Diagnosis

Isolated hemangioblastoma, retinal angioma, or clear cell renal cell carcinoma. The clinical sensitivity of molecular genetic testing of VHL makes it possible to effectively rule out von Hippel-Lindau syndrome (VHL) with a high degree of certainty in individuals with (1) isolated hemangioblastoma, retinal angioma, or clear cell renal cell carcinoma and (2) no detectable germline VHL pathogenic variant. Somatic mosaicism for a VHL pathogenic variant could still be considered in such individuals (an estimated 5% of individuals with VHL have a somatic mosaicism [Chen et al 2022]). A younger individual, especially one with multiple lesions, is more likely to have a germline VHL pathogenic variant than an older individual with a single lesion [Binderup et al 2017a].

The differential diagnosis for an individual suspected of having VHL depends on the further clinical presentation (see Table 3a and Table 3b).

Pheochromocytoma. Up to 40% of individuals with pheochromocytoma have a heterozygous pathogenic variant in one of several genes: SDHB, SDHD, VHL, RET, and NF1 (most prevalent) or SDHA, SDHAF2, MAX, FH, and TMEM127 (less prevalent). Germline VHL pathogenic variants are rare in simplex cases of unilateral pheochromocytoma (i.e., an affected individual with no family history of VHL), unless the individual is younger than age 20 years.

Table 3a.

Table 3a.

Genetic Disorders Associated with an Increased Risk of Pheochromocytomas

Renal cell carcinoma. Individuals with familial renal cell carcinoma should be evaluated for FH tumor predisposition syndrome and Birt-Hogg-Dubé syndrome.

Table 3b.

Table 3b.

Genetic Disorders Associated with Renal Cell Carcinoma

Endolymphatic sac tumors in VHL are often misdiagnosed as Ménière disease.

Management

The first von Hippel-Lindau syndrome (VHL) care pathway was recently published by a multidisciplinary team from the Netherlands [Wolters et al 2022].

Evaluations Following Initial Diagnosis

To establish the extent of disease and needs in an individual diagnosed with VHL, the evaluations summarized in Table 4 (if not performed as part of the evaluation that led to the diagnosis) are recommended.

Table 4.

Table 4.

Von Hippel-Lindau Syndrome: Recommended Evaluations Following Initial Diagnosis

Treatment of Manifestations

Targeted Therapies

In GeneReviews, a targeted therapy is one that addresses the specific underlying mechanism of disease causation (regardless of whether the therapy is significantly efficacious for one or more manifestation of the genetic condition); would otherwise not be considered without knowledge of the underlying genetic cause of the condition; or could lead to a cure. —ED

Pazopanib is an FDA-approved treatment for advanced renal cell carcinoma. A single-arm Phase II trial included 31 individuals with a molecular or clinical diagnosis of VHL treated with 800 mg oral pazopanib for 24 weeks [Giles & Gläsker 2018, Jonasch et al 2018]. Four of the individuals were negative for a germline VHL pathogenic variant. Thirteen of 31 (42%) individuals achieved a response based on RECIST criteria after treatment with pazopanib. More than half (52%) of individuals with renal cell carcinoma responded to therapy, including two individuals with complete response. Similarly, 52% of pancreatic lesions showed partial or complete response and two of 49 hemangioblastomas showed partial response. One individual died due to hemorrhage of a central nervous system (CNS) hemangioma. Response to treatment was limited to individuals with a germline pathogenic variant. The study concluded that systemic treatment with pazopanib can be considered in individuals with a germline VHL pathogenic variant and without a CNS hemangioblastoma requiring clinical action.

Belzutifan. Promising results were demonstrated in the LITESPARK-004 trial of hypoxia-inducible factor 2 alpha (HIF2α) inhibitor belzutifan in individuals with VHL and clear cell renal cell carcinoma. Belzutifan is a targeted oral therapy specifically altering the biology of cells lacking the VHL protein [Choueiri et al 2021]. Belzutifan has only limited side effects. The approval was based on a Phase II clinical trial following 61 individuals with VHL that reported clinical benefit for individuals with all tumor types, including a drastic reduction in the number of required surgeries [Jonasch et al 2021]. A recent update of nearly 2.5 years of follow up of the 61 individuals reported that belzutifan continued to show anti-tumor activity in VHL-related neoplasms, including tumors in the kidney, pancreas, brain, spine, and eyes, with no new safety concerns [Jonasch et al 2022]. Belzutifan induced partial responses with a renal cell carcinoma objective response rate (ORR) of 49% and a disease control rate of 98.4% after 21.8 months of treatment. All individuals with pancreatic lesions (e.g., cysts, pancreatic neuroendocrine tumors) had an ORR of 77%, and those with CNS hemangioblastoma had a 30% response rate. In total, 33% had grade 3 or higher adverse events reported, and seven individuals (11.5%) discontinued the treatment. Belzutifan was FDA approved in August 2021, and is used in individuals not requiring immediate surgery. It is unknown if belzutifan can prevent tumors in individuals with VHL. The most prominent side effects include anemia and fatigue.

Supportive Care

No guidelines exist for the management of VHL lesions.

CNS hemangioblastomas

  • Most CNS hemangioblastomas can be surgically removed completely and safely [Gläsker et al 2013]. Intraoperative videoangiography, ultrasound, and fluorescein optimize identification of CNS hemangioblastomas and their vasculature, leading to a safer resection with reduced side effects [Mazzapicchi et al 2022]. Intraoperative fluorescence angiography was helpful in reducing intraoperative bleeding and preventing spinal swelling [Mehta et al 2017].
  • Some advocate early surgical removal of both symptomatic and asymptomatic CNS hemangioblastomas, while others follow asymptomatic lesions with yearly imaging studies. A study of 15 symptomatic individuals with cauda equina hemangioblastomas revealed a worse outcome in only one individual six months after surgery. The other individuals were stable or improved [Mehta et al 2017].

Spinal hemangioblastomas

  • Preoperative arterial embolization may be indicated, especially for extensive spinal tumors. A minimally invasive approach for the resection of selected spinal hemangioblastomas is considered safe and allows complete tumor resection [Krüger et al 2019].
  • Pathologic findings during intraoperative neurophysiologic monitoring (IONM) appear to predict worse long-term outcomes after microsurgical removal of spinal cord hemangioblastomas [Siller et al 2017]. The use of IONM seems to be associated with better neurologic outcomes [Feletti et al 2022]. Spinal cord surgery should be performed in an expert center by experienced neurosurgeons.
  • Surgical intervention for cysts/syrinx in the spinal cord is recommended.
  • Stereotactic therapy is increasingly popular, but there is still a need for prospective studies [Pan et al 2018]. Gamma knife surgery may be useful with small solid tumors or those in inoperable sites [Asthagiri et al 2010, Simone et al 2011]. While this technique may reduce the size of the solid tumor, it does not appear to prevent cyst formation. The unpredictable growth pattern makes it difficult to determine when to start stereotactic therapy in order to avoid unnecessary intervention. Recent studies show promising local control rates at one, three, and five years were 96%, 92%, and 92%, respectively. Clinically, 13/16 (81.2%) tumors had symptomatic improvement [Pan et al 2017].
  • Factors associated with tumor control are solid, smaller tumors, VHL-associated lesions, and higher margin dose. Thirteen of 186 individuals (7%) experienced complications, 11 individuals needed steroid therapy, and one person died of refractory peritumoral edema. Two individuals required additional surgery [Kano et al 2015].
  • Another study showed recurrence-free survival in six of eight individuals at a mean follow up of 48 months. Two individuals required additional surgery for persisting cerebellar symptoms. One individual showed an increase in cyst volume along with a decrease of the size of the mural nodule [Goyal et al 2016].
  • A case study showed complete loss of stromal cells after a standard dose of stereotactic radiosurgery for hemangioblastoma, indicating the effectiveness of the treatment [Nambu et al 2018].

Retinal hemangioblastomas

  • Ultra-widefield fluorescein angiography can be useful in the evaluation and management of retinal hemangioblastomas. This technique appears to detect more hemangioblastomas than ophthalmoscopy and conventional angiography [Chen et al 2018].
  • Most ophthalmologists favor prospective treatment of retinal hemangioblastomas (but not optic nerve hemangioblastomas) to avoid blindness, although spontaneous regression has occurred.
  • Therapeutic modalities used to treat retinal hemangioblastomas include diathermy, xenon, laser, and cryocoagulation, with variable degrees of success depending on the location, size, and number of lesions. Recurrent tumors have been noted, even after many years, but some may be new tumors in the same general area rather than recurrent disease.
  • Vitreoretinal surgery is indicated when retinal hemangioblastomas are accompanied by complications such as epiretinal membrane development, vitreous hemorrhage, tractional and/or exudative retinal detachment, preretinal fibrosis, or proliferative vitreoretinopathy [Gaudric et al 2011].
  • Brachytherapy could be a treatment option for larger peripheral hemangioblastomas with manageable risk and a high eye preservation rate [Dalbah et al 2021].
  • External beam radiotherapy has been shown to be useful when standard therapy has not prevented progression [Raja et al 2004]. In individual reports early surgical resection and intravitreal treatment with bevacizumab and propranolol were considered safe and effective [Agarwal et al 2016, van Overdam et al 2017, Karimi et al 2020].
  • There is no evidence to support the use of sunitinib for retinal hemangioblastomas.

Renal cell carcinoma

  • Local therapies. Image-guided techniques such as cryoablation and radiofrequency ablation are now preferred over surgical resection for small renal cell carcinomas [Chan et al 2022]; outcomes are excellent and the procedures are considered safe [Allasia et al 2017, Carrion et al 2020, Wessendorf et al 2021, Chan et al 2022]. Radiofrequency ablation therapy is often applied to smaller tumors, particularly <3 cm [Best et al 2012, Carrion et al 2020]. The major complication rate resulting in a need for a radiologic, surgical, or endoscopic intervention following laparoscopic and percutaneous radiofrequency ablation therapy was 7.3% and 4.3%, respectively [Young et al 2012], but other studies showed no or only minor complications [Allasia et al 2017, Wessendorf et al 2021].
  • In individuals treated with nephrectomy, the adrenal gland should be left in situ. If contralateral pheochromocytoma occurs requiring adrenalectomy, the remaining adrenal gland will prevent or delay adrenal insufficiency.
  • Kidney transplantation has been successful in individuals in whom bilateral nephrectomy has been necessary. It is imperative to evaluate any living related potential donor for VHL and to exclude those with VHL.

Pheochromocytomas

  • Pheochromocytomas should be surgically removed. Laparoscopic approaches have been shown to be effective and safe [Dickson et al 2011, Agarwal et al 2012]. It has been proposed that active surveillance for pheochromocytomas smaller than 2 cm can be safe [Sanford et al 2021].
  • Preoperative treatment with alpha-adrenergic blockade and optional additional beta-adrenergic blockade for seven to ten days is appropriate even in individuals with no known hypertension and is recommended in treatment guidelines. However, perioperative alpha-adrenergic blockade has recently been under debate. In a multicenter study, mortality rate in pretreated individuals was slightly higher than in non-pretreated individuals [Groeben et al 2020].
  • Adrenal-sparing surgery should be considered and is regarded as a successful treatment strategy. Studies show low recurrence, no metastatic disease, and rare steroid dependency after cortical-sparing techniques [Sanford et al 2021, Shirali et al 2023]. Moreover, survival is not affected by adrenal-sparing surgery [Neumann et al 2019]. Adrenal-sparing surgery is the therapy of choice in children. In ten individuals with VHL, 18 successful operations were performed. After follow up (median 7.2 years), two individuals developed a new tumor in the ipsilateral adrenal gland [Volkin et al 2012].

Pancreatic cysts and neuroendocrine tumors

  • Pancreatic cysts are common, rarely influence endocrine function, and have no malignant behavior. Therefore, surgical removal is not generally required [Sharma et al 2017].
  • Pancreatic neuroendocrine tumors (PNETs) need to be differentiated from cysts and serous cystadenomas. PNETs are generally slow growing and are not hormonally active, although they can cause metastatic disease. Surgical resection should be strongly considered when there is a high risk of metastases, as suggested by one of the following prognostic criteria: (1) size ≥2.5 cm or (2) tumor doubling rate <500 days [Krauss et al 2018]. Individuals with a VHL missense variant are reported to have a higher rate of metastases with PNET size between 1.2-3.0 cm in comparison to individuals with other types of VHL pathogenic variants [Tirosh et al 2018b].

Endolymphatic sac tumors (ELST). Consideration of surgical removal of these slow-growing tumors must include discussion of the possible complication of total deafness. Early intervention with small tumors has been shown to preserve both hearing and vestibular function [Friedman et al 2013]. Friedman et al [2013] described two individuals (2/18) with postoperative decreased facial nerve function and three (3/18) individuals with recurrent ELSTs (with a mean follow up of 67 months). In 31 individuals with VHL with 33 resected ELSTs, 29 individuals were symptomatic. After surgery, hearing was stabilized or improved in 97% of individuals, and tumor resection was complete in 91%. Complications occurred in three tumors: cerebrospinal fluid leakage in two (6%) and transient lower cranial nerve palsy in one (3%) [Kim et al 2013].

Epididymal or broad ligament papillary cyst adenomas generally do not require surgical resection unless they are symptomatic or are threatening fertility.

Surveillance

In the United States, the VHL Alliance has worked extensively with health care professionals to assemble guidelines that are generally accepted worldwide (see VHL Resources – Active Surveillance Guidelines). Additional guidelines originate from Denmark and the Netherlands [Wolters et al 2022]. Individuals with VHL and first-degree relatives who have not undergone DNA-based testing need regular clinical surveillance by a physician or medical team familiar with VHL (see Table 5).

Table 5.

Table 5.

Von Hippel-Lindau Syndrome: Recommended Surveillance

Agents/Circumstances to Avoid

Avoid the following:

  • Tobacco products, as they are considered a risk factor for kidney cancer
  • Chemicals and industrial toxins known to affect VHL-involved organs
  • Contact sports if adrenal or pancreatic lesions are present

Evaluation of Relatives at Risk

Early recognition of manifestations of VHL may allow for timely intervention and improved outcome; thus, clinical surveillance of asymptomatic at-risk individuals (including children) for early manifestations of VHL is appropriate. The American Society of Clinical Oncology identifies VHL as a Group 1 disorder – a hereditary disease for which genetic testing is considered part of the standard management for at-risk family members [Robson et al 2015].

  • If the VHL pathogenic variant in the family is known, molecular genetic testing can be used for early identification of at-risk family members to improve diagnostic certainty and reduce the need for screening procedures in those at-risk family members who have not inherited the pathogenic variant [Priesemann et al 2006].
  • If the VHL pathogenic variant in the family is not known and/or at-risk individuals decline genetic testing, continued screening for VHL lesions is warranted (see Surveillance).

The use of molecular genetic testing for determining the genetic status of presumably at-risk relatives when a family member with a clinical diagnosis of VHL is not available for testing is not straightforward. Such test results need to be interpreted with caution. A positive test result signals the presence of a VHL pathogenic variant in the at-risk family member and indicates that the same molecular genetic testing method can be used to assess the genetic status of other at-risk family members. However, a negative test for a VHL pathogenic variant in an at-risk family member under such circumstances suggests one of the following possibilities:

  • The at-risk family member has not inherited a VHL pathogenic variant.
  • The familial VHL pathogenic variant may not be detectable by the assays used.
  • The clinical diagnosis of VHL in the proband is questionable.

In this situation, the presumably at-risk family member has a small but finite residual risk of having inherited a pathogenic allele (i.e., VHL or other hereditary disorder). In counseling such individuals, careful consideration should be given to the strength of the clinical diagnosis of VHL in the affected family member, the relationship of the at-risk individual to the affected family member, the perceived risk of an undetected VHL (or other gene) pathogenic variant, and the potential need for some form of continued clinical surveillance.

See Genetic Counseling for issues related to testing of at-risk relatives for genetic counseling purposes.

Pregnancy Management

There is no consensus regarding medical surveillance for pregnant women with VHL. Research by the French VHL Study Group showed a significantly higher complication rate of hemangioblastomas in individuals with VHL who had had at least one pregnancy [Abadie et al 2010]. Another study concluded that pregnancy has a significant influence on cerebellar hemangioblastoma growth and causes an overall high complication rate (17%) [Frantzen et al 2012]. Intensified surveillance can be considered in a specialized medical center prior to conception and during pregnancy to allow early identification of pheochromocytomas and cerebellar hemangioblastomas. A recent study showed a decrease in new VHL manifestations during pregnancy [Binderup et al 2015]. In another study pregnancy did not correlate with the development of new hemangioblastomas or hemangioblastoma or cyst growth [Ye et al 2012], suggesting that more frequent surveillance is not needed during pregnancy. The VHL Active Surveillance Guidelines recommends MRI of the cerebellum without contrast at four months' gestation.

See MotherToBaby for further information on medication use during pregnancy.

Therapies Under Investigation

Certain VHL pathogenic variants fail to downregulate hypoxia-inducible factor alpha (HIFα), leading to overexpression of downstream effectors such as vascular endothelial growth factor (VEGF), which contribute to pathogenesis. Many experimental therapies target these misregulated signaling pathways. An intravitreal VEGF receptor inhibitor, ranibizumab, has been used with some success in individuals with retinal hemangioblastomas who have either failed local therapy or whose lesions are not amenable to local therapy [Wong et al 2008]. Intravitreal injections of bevacizumab, another VEGF inhibitor, have also proven effective in treating retinal hemangioblastomas in individuals with VHL [Hrisomalos et al 2010]. Stabilization of some (but not all) CNS hemangioblastomas has also been demonstrated [Madhusudan et al 2004].

Sunitinib, a tyrosine kinase inhibitor (TKI) that inhibits the action of VEGF receptors, has had some utility in the rare unresectable malignant pheochromocytomas, but simple surgical excision is clearly preferable for these usually benign tumors [Jimenez et al 2009]. Sunitinib has also been shown to effectively treat clear cell renal cell carcinomas – but not hemangioblastomas – in individuals with VHL [Jonasch et al 2011].

Pazopanib showed favorable effects on the clinical condition of individuals with recurrent and rapidly progressive VHL-associated hemangioblastomas [Migliorini et al 2015]. A pilot study to assess the safety and efficacy of another TKI, dovitinib, for the treatment of asymptomatic hemangioblastomas in individuals with VHL resulted in termination of the study after adverse events in all six individuals. Maculopapular rash, diarrhea, and fatigue were most common [Pilié et al 2018]. In a series of 22 individuals with VHL with a total of 311 lesions, good identification of VEGF-producing lesions suggests that 89Zr-bevacizumab PET could offer a tool to select individuals for anti-VEGF therapy [Oosting et al 2016].

Somatostatin analogs could be of use in the treatment of hemangioblastomas. Nine hemangioblastomas demonstrated expression for at least three somatostatin receptor subtypes (1, 2a, 3, 4, or 5). One individual with a symptomatic irresectable suprasellar hemangioblastoma was treated with octreotide long-acting release, which resulted in clinical stability and radiographic response after nine months of treatment [Sizdahkhani et al 2017].

Propranolol could be an efficient treatment to control hemangioblastoma growth in individuals with VHL because of its antiangiogenic effects demonstrated in infantile hemangioma and the hypothetical impact on HIF levels.

Checkpoint inhibitors such as antibodies targeting programmed cell death ligand 1 (PD-L1) have shown promise in managing tumor load; however, these treatments have unknown toxicity in individuals with VHL, who will likely have dozens to thousands of small subclinical lesions present throughout their body.

Sardi et al [2009] reported three-year stabilization of previously progressive multifocal spinal hemangioblastomas with thalidomide.

Premature termination codon 124 (PTC124), also known as ataluren, may benefit a subset of affected individuals in whom nonsense variants give rise to premature stop codons in the messenger RNA [Auld et al 2010]. There are three stop codons: UAA, UAG, and UGA. PTC124 promotes read-through of all three stop codons with different efficiencies. The highest read-through efficiency takes place at UGA, followed by UAG and then UAA. PTC124 has been successfully proven to promote read-through of nonsense variants in Duchenne muscular dystrophy (DMD), cystic fibrosis (CF), and Usher syndrome type 1C. Phase I and II clinical trials have shown no serious side effects with PTC124 treatment, even after long-term use [Wilschanski et al 2011]. Preclinical investigation of PTC124 effects on VHL is ongoing.

An in vivo study of HIF2α inhibitor in vhl-/- zebra fish showed promising results in suppressing erythrocytosis and abnormal vascular proliferation in the brain and trunk. Furthermore, it promoted erythroid differentiation and decreased the number of early erythroid progenitors circulating in the peripheral blood. Therefore, there is a rationale for performing preclinical and clinical studies in optimized HIF2α inhibitors [Metelo et al 2015].

Search ClinicalTrials.gov in the US and EU Clinical Trials Register in Europe for access to information on clinical studies for a wide range of diseases and conditions.

Genetic Counseling

Genetic counseling is the process of providing individuals and families with information on the nature, mode(s) of inheritance, and implications of genetic disorders to help them make informed medical and personal decisions. The following section deals with genetic risk assessment and the use of family history and genetic testing to clarify genetic status for family members; it is not meant to address all personal, cultural, or ethical issues that may arise or to substitute for consultation with a genetics professional. —ED.

Mode of Inheritance

Von Hippel-Lindau syndrome (VHL) is inherited in an autosomal dominant manner.

Risk to Family Members

Parents of a proband

  • Approximately 80% of individuals diagnosed with VHL have an affected parent.
  • Some individuals diagnosed with VHL have the disorder as the result of a VHL pathogenic variant that occurred as a de novo event in the affected individual or as a postzygotic de novo event in a mosaic, apparently unaffected parent. The proportion of individuals with VHL due to a de novo pathogenic variant is about 20%.
  • If the proband appears to be the only affected family member:
    • And a molecular diagnosis has been established in the proband, genetic testing is recommended for the parents of the proband to confirm their genetic status and to allow reliable recurrence risk counseling.
    • And a molecular diagnosis has not been established in the proband, ophthalmologic screening and abdominal ultrasound evaluation, at a minimum, should be offered to both parents.
    Note: Screening for VHL lesions is warranted for parents without clinical manifestations but who are identified as having a VHL pathogenic variant and for parents who have not undergone DNA-based testing (see Surveillance).
  • If the pathogenic variant identified in the proband is not identified in either parent and parental identity testing has confirmed biological maternity and paternity, the following possibilities should be considered:
    • The proband has a de novo pathogenic variant.
    • The proband inherited a pathogenic variant from a parent with germline (or somatic and germline) mosaicism* [Murgia et al 2000, Sgambati et al 2000, Santarpia et al 2007, Wu et al 2013, Coppin et al 2014, Chen et al 2022]. Note: Testing of parental leukocyte DNA may not detect all instances of somatic mosaicism and will not detect a pathogenic variant that is present in the germ cells only.
      * A parent with somatic and germline mosaicism for a VHL pathogenic variant may be mildly/minimally affected.
  • The family history of some individuals diagnosed with VHL may appear to be negative because of failure to recognize the disorder in family members, reduced penetrance, early death of the parent before the onset of symptoms, or late onset of the syndrome in the affected parent. Therefore, an apparently negative family history cannot be confirmed unless molecular genetic testing has demonstrated that neither parent is heterozygous for the pathogenic variant identified in the proband.

Sibs of a proband. The risk to the sibs of the proband depends on the clinical/genetic status of the proband's parents:

  • If a parent of the proband is affected and/or is known to have the VHL pathogenic variant, the risk to the sibs of inheriting the pathogenic variant is 50%. The degree of clinical severity is not predictable in sibs who inherit a VHL pathogenic variant, as the manifestations and severity of VHL can be highly variable among family members with the same pathogenic variant.
  • If the proband has a known VHL pathogenic variant that cannot be detected in the leukocyte DNA of either parent, the recurrence risk to sibs is slightly greater than that of the general population because of the possibility of parental germline mosaicism. Parental mosaicism has been described. In families in which the proband represents a simplex case, the incidence of parental mosaicism is thought to be about 5% [Chen et al 2022].
  • If the genetic status of the parents is unknown but the parents are clinically unaffected and are at least age 35 years, the risk to the sibs of a proband appears to be low; however, the sibs are still at increased risk for VHL because of the possibility of failure to recognize the disorder or late onset of the syndrome in an affected parent and the possibility of parental germline mosaicism.

Offspring of a proband. Each child of an individual with VHL is at a 50% risk of inheriting the VHL pathogenic variant; the degree of clinical severity is not predictable.

Other family members. The risk to other family members depends on the status of the proband's parents: if a parent is affected and/or has the VHL pathogenic variant, the parent's family members may be at risk.

Related Genetic Counseling Issues

See Management, Evaluation of Relatives at Risk for information on evaluating at-risk relatives for the purpose of early diagnosis and treatment.

Genetic cancer risk assessment and counseling. For a comprehensive description of the medical, psychosocial, and ethical ramifications of identifying at-risk individuals through cancer risk assessment with or without molecular genetic testing, see Cancer Genetics Risk Assessment and Counseling – for health professionals (part of PDQ®, National Cancer Institute).

Testing of at-risk asymptomatic family members. Molecular genetic testing of at-risk family members is appropriate in order to determine the need for continued clinical surveillance. Interpretation of molecular genetic test results is most accurate when a germline VHL pathogenic variant has been identified in an affected family member (see Evaluation of Relatives at Risk).

Because early detection of at-risk individuals affects medical management, testing of asymptomatic individuals during childhood is beneficial [Binderup et al 2022] (see VHL Resources – VHL Patient and Caregiver Handbook). As ophthalmologic screening for those at risk for VHL begins as early as possible, certainly before age five years, molecular genetic testing may be considered in young children. Molecular genetic testing may be performed earlier if the results would alter the medical management of the child.

Parents often want to know the genetic status of their children prior to initiating screening in order to avoid unnecessary procedures in a child who has not inherited the pathogenic variant. Special consideration should be given to education of the children and their parents prior to genetic testing. A plan should be established for the manner in which results are to be given to the parents and their children. The authors recommend the VHL Kids Handbook by the VHL Alliance (see VHL Resources – VHL Kids Handbook).

Other issues to consider. It is recommended that physicians ordering VHL molecular genetic testing and individuals considering undergoing testing understand the risks, benefits, and limitations of the testing prior to sending a sample to a laboratory. Referral to a genetic counselor and/or a center in which testing is routinely offered is recommended.

Family planning

  • The optimal time for determination of genetic risk and discussion of the availability of prenatal/preimplantation genetic testing is before pregnancy.
  • It is appropriate to offer genetic counseling (including discussion of potential risks to offspring and reproductive options) to young adults who are affected or at risk.
  • Preconception considerations include possible male infertility due to cysts of the epididymis.

DNA banking. Because it is likely that testing methodology and our understanding of genes, pathogenic mechanisms, and diseases will improve in the future, consideration should be given to banking DNA from probands in whom a molecular diagnosis has not been confirmed (i.e., the causative pathogenic mechanism is unknown). For more information, see Huang et al [2022].

Prenatal Testing and Preimplantation Genetic Testing

Once the VHL pathogenic variant has been identified in an affected family member, prenatal and preimplantation genetic testing for VHL are possible.

Differences in perspective may exist among medical professionals and in families regarding the use of prenatal testing. While most centers would consider use of prenatal testing to be a personal decision, discussion of these issues may be helpful.

Resources

GeneReviews staff has selected the following disease-specific and/or umbrella support organizations and/or registries for the benefit of individuals with this disorder and their families. GeneReviews is not responsible for the information provided by other organizations. For information on selection criteria, click here.

Molecular Genetics

Information in the Molecular Genetics and OMIM tables may differ from that elsewhere in the GeneReview: tables may contain more recent information. —ED.

Table A.

Table A.

Von Hippel-Lindau Syndrome: Genes and Databases

Table B.

Table B.

OMIM Entries for Von Hippel-Lindau Syndrome (View All in OMIM)

Molecular Pathogenesis

The role of von Hippel-Lindau disease tumor suppressor (pVHL) in the regulation of hypoxia-inducible genes through the targeted ubiquitinylation and degradation of hypoxia-inducible factor 1 alpha (HIF1α) has been described in great detail and won the 2019 Nobel Prize, leading to a model of how disruption of VHL results in renal cell carcinoma, hemangioblastoma, and the production of other highly vascularized tumors [Takamori et al 2023].

Von Hippel-Lindau syndrome (VHL) results from a germline loss-of-function variant coupled with a somatic loss-of-function variant involving the second allele. Pathogenic variants can prevent or reduce VHL expression or lead to the expression of an abnormal protein. Some genotype-phenotype correlations are observed [Reich et al 2021]. Pathogenic missense variants that destabilize packing of the alpha-helical domains, decrease the stability of the alpha-beta domain interface, interfere with binding of elongin C and HIF1α, or disrupt hydrophobic core residues result in loss of HIF regulation and are more likely to result in VHL type 1. Pathogenic missense variants that do not disrupt HIF regulation are more likely to be associated with VHL type 2. Furthermore, VHL pathogenic variants affect vessel branching and maturation via the Notch signaling pathway [Arreola et al 2018].

Pathogenic missense variants that lead to pheochromocytoma with a low (or no) risk for renal cell carcinoma (VHL types 2A and 2C) may encode pVHL that retains the ability to ubiquinate (and thereby downregulate) HIF1α in the presence of molecular oxygen to a greater degree than pathogenic variants that result in VHL with pheochromocytoma and renal cell carcinoma (VHL type 2B). Furthermore, mutated pVHL may predispose to pheochromocytoma by altering the balance among a group of proteins in a molecular pathway that controls apoptosis of sympatho-adrenal precursor cells during development.

Mechanism of disease causation. Loss of function

VHL-specific laboratory technical considerations. Sequence analysis of intron 1 should be performed in probands without an identified VHL pathogenic variant in the coding region, as some intron 1 pathogenic variants can lead to inclusion of a cryptic exon (designated exon E1') [Lenglet et al 2018].

Chapter Notes

Author History

Saya Ahmad, BSc (2018-present)
Debra L Collins, MS; University of Kansas Medical Center (2000-2012)
Carlijn Frantzen, MD; University Medical Center Groningen (2012-2018)
Timothy Klasson, BSc; University Medical Center Utrecht (2015-2018)
Rachel Giles, MD, PhD (2012-present)
Thera P Links, MD, PhD; University Medical Center Groningen (2012-2023)
R Neil Schimke, MD; University of Kansas Medical Center (2000-2012)
Catherine A Stolle, PhD; The Children's Hospital of Philadelphia (2000-2012)
Rachel S van Leeuwaarde, MD (2018-present)
Bernadette van Nesselrooij, MD, PhD (2023-present)
Wouter Zandee, MD, PhD (2023-present)

Revision History

  • 29 February 2024 (aa) Revision: clinical diagnostic criteria in Table 1 updated
  • 21 September 2023 (sw) Comprehensive update posted live
  • 6 September 2018 (sw) Comprehensive update posted live
  • 6 August 2015 (me) Comprehensive update posted live
  • 21 June 2012 (me) Comprehensive update posted live
  • 22 December 2009 (me) Comprehensive update posted live
  • 20 March 2007 (me) Comprehensive update posted live
  • 1 December 2004 (me) Comprehensive update posted live
  • 14 November 2002 (tk,cg) Comprehensive update posted live
  • 17 May 2000 (me) Review posted live
  • 17 September 1999 (rns) Original submission

References

Published Guidelines / Consensus Statements

  • American Society of Clinical Oncology. Policy statement update: genetic testing for cancer susceptibility. 2003.
  • American Society of Clinical Oncology. Policy statement update: genetic testing for cancer susceptibility. 2010.
  • Binderup MLM, Smerdel M, Borgwadt L, Beck Nielsen SS, Madsen MG, Møller HU, Kiilgaard JF, Friis-Hansen L, Harbud V, Cortnum S, Owen H, Gimsing S, Friis Juhl HA, Munthe S, Geilswijk M, Rasmussen ÅK, Møldrup U, Graumann O, Donskov F, Grønbæk H, Stausbøl-Grøn B, Schaffalitzky de Muckadell O, Knigge U, Dam G, Wadt KA, Bøgeskov L, Bagi P, Lund L, Stochholm K, Ousager LB, Sunde L. Von Hippel-Lindau disease: Updated guideline for diagnosis and surveillance. Eur J Med Genet. 2022;65:104538. [PubMed]
  • Hes FJ, van der Luijt RB, Lips CJ. Clinical management of von Hippel-Lindau (VHL) disease. Neth J Med. 2001;59:225-34. [PubMed]
  • Lonser RR, Glenn GM, Walther M, Chew EY, Libutti SK, Linehan WM, Oldfield EH. Von Hippel-Lindau disease. Lancet. 2003;361:2059-67. [PubMed]
  • VHL Alliance. VHL Patient and Caregiver Handbook. Available online. 2020. Accessed 8-28-23.

Literature Cited

  • Abadie C, Croupier I, Bringuier-Branchereau S, Mercies G, Deveaux S, Richard S. Role of pregnancy on hemangioblastomas in von Hippel-Lindau disease: a retrospective French study. Rio de Janeiro, Brazil: 9th International Medical Symposium on VHL. 2010.

  • Abbott MA, Nathanson KL, Nightingale S, Maher ER, Greenstein RM. The von Hippel-Lindau (VHL) germline mutation V84L manifests as early-onset bilateral pheochromocytoma. Am J Med Genet A. 2006;140:685–90. [PubMed: 16502427]

  • Agarwal A, Kumari N, Singh R. Intravitreal bevacizumab and feeder vessel laser treatment for a posteriorly located retinal capillary hemangioma. Int Ophthalmol. 2016;36:747-50. [PubMed: 26815929]

  • Agarwal G, Sadacharan D, Aggarwal V, Chand G, Mishra A, Agarwal A, Verma AK, Mishra SK. Surgical management of organ-contained unilateral pheochromocytoma: comparative outcomes of laparoscopic and conventional open surgical procedures in a large single-institution series. Langenbecks Arch Surg. 2012;397:1109-16. [PubMed: 22120010]

  • Allasia M, Soria F, Battaglia A, Gazzera C, Calandri M, Caprino MP, Lucatello B, Velrti A, Maccario M, Pasini B, Bosio A, Gontero P, Destefanis P. Radiofrequency ablation for renal cancer in Von Hippel-Lindau syndrome patients: a prospective cohort analysis. Clin Genitourin Cancer. 2017;S1558-7673:30237-9. [PubMed: 28866246]

  • Arreola A, Payne LB, Julian MH, de Cubas AA, Daniels AB, Taylor S, Zhao H, Darden J, Bautch VL, Rathmell WK, Chappell JC. Von Hippel-Lindau mutations disrupt vascular patterning and maturation via Notch. JCI Insight. 2018;3:e92193. [PMC free article: PMC5916240] [PubMed: 29467323]

  • Asthagiri AR, Metha GU, Zach L, Li X, Butman JA, Camphausen KA, Lonser RR. Prospective evaluation of radiosurgery for hemangioblastomas in von Hippel-Lindau disease. Neuro Oncol. 2010;12:80-6. [PMC free article: PMC2940550] [PubMed: 20150370]

  • Auld DS, Lovell S, Thorne N, Lea WA, Maloney DJ, Shen M, Rai G, Battaile KP, Thomas CJ, Simeonov A, Hanzlik RP, Inglese J.) Molecular basis for the high-affinity binding and stabilization of firefly luciferase by PTC124. Proc Natl Acad Sci U S A. 2010;107:4878-83. [PMC free article: PMC2841876] [PubMed: 20194791]

  • Bento C. Genetic basis of congenital erythrocytosis. Int J Lab Hematol. 2018;40:62-67. [PubMed: 29741264]

  • Bertelsen M, Kosteljanetz M. An evaluation of Danish national clinical guidelines for von Hippel-Lindau (VHL). Acta Neurochir (Wien). 2011;153:35-41. [PubMed: 20865287]

  • Best SL, Park SK, Youssef RF, Olweny EO, Tan YK, Trimmer C, Cadeddu JA. Long-term outcomes of renal tumor radio frequency ablation stratified by tumor diameter: size matters. J Urol. 2012;187:1183-9. [PubMed: 22335865]

  • Binderup ML, Budtz-Jørgensen E, Bisgaard ML. New von Hippel-Lindau manifestations develop at the same or decreased rates in pregnancy. Neurology. 2015;85:1500-3. [PubMed: 26408493]

  • Binderup ML, Budtz-Jørgensen E, Bisgaard ML. Risk of new tumors in von Hippel-Lindau patients depends on age and genotype. Genet Med. 2016;18:89-97. [PubMed: 25834951]

  • Binderup ML, Galanakis M, Budtz-Jørgensen E, Kosteljanetz M, Luise Bisgaard M. Prevalence, birth incidence, and penetrance of von Hippel-Lindau disease (VHL) in Denmark. Eur J Hum Genet. 2017a;25:301-7. [PMC free article: PMC5315510] [PubMed: 27966541]

  • Binderup ML, Gimsing S, Kosteljanetz M, Thomsen C, Bisgaard ML. von Hippel-Lindau disease: deafness due to a non-MRI-visible endolymphatic sac tumor despite targeted screening. Int J Audiol. 2013;52:771-5. [PubMed: 24003980]

  • Binderup ML, Jensen AM, Budtz-Jørgensen E, Bisgaard ML. Survival and causes of death in patients with von Hippel-Lindau disease. J Med Genet 2017b;54:11-18. [PubMed: 27539272]

  • Binderup MLM, Smerdel M, Borgwadt L, Beck Nielsen SS, Madsen MG, Møller HU, Kiilgaard JF, Friis-Hansen L, Harbud V, Cortnum S, Owen H, Gimsing S, Friis Juhl HA, Munthe S, Geilswijk M, Rasmussen ÅK, Møldrup U, Graumann O, Donskov F, Grønbæk H, Stausbøl-Grøn B, Schaffalitzky de Muckadell O, Knigge U, Dam G, Wadt KA, Bøgeskov L, Bagi P, Lund L, Stochholm K, Ousager LB, Sunde L. Von Hippel-Lindau disease: updated guideline for diagnosis and surveillance. Eur J Med Genet. 2022;65:104538. [PubMed: 35709961]

  • Boedeker CC, Hensen EF, Neumann HP, Maier W, van Nederveen FH, Suárez C, Kunst HP, Rodrigo JP, Takes RP, Pellitteri PK, Rinaldo A, Ferlito A. Genetics of hereditary head and neck paragangliomas. Head Neck. 2014;36:907-16. [PubMed: 23913591]

  • Bond E, Yashar B, Else T, Osborne J, Marvin M. Disclosure of genetic risk to dating partners among young adults with von Hippel-Lindau disease. Fam Cancer. 2023;22:203-15. [PubMed: 35984582]

  • Butman JA, Nduom E, Kim HJ, Lonser RR. Imaging detection of endolymphatic sac tumor-associated hydrops. J Neurosurg. 2013;119:406-11. [PubMed: 23472846]

  • Carrion DM, Linares-Espinós E, Ríos González E, Bazán AA, Alvarez-Maestro M, Martinez-Pineiro L. Invasive management of renal cell carcinoma in von Hippel-Lindau disease. Cent European J Urol. 2020;73:167-72. [PMC free article: PMC7407786] [PubMed: 32782836]

  • Catapano D, Muscarella LA, Guarnieri V, Zelante L, D'Angelo VA, D'Agruma L. Hemangioblastomas of central nervous system: molecular genetic analysis and clinical management. Neurosurgery. 2005;56:1215-21. [PubMed: 15918937]

  • Chan VW, Lenton J, Smith J, Jagdev S, Ralph C, Vasudev N, Bhattarai S, Lewington A, Kimuli M, Cartledge J, Wah TM. Multimodal image-guided ablation on management of renal cancer in Von-Hippel-Lindau syndrome patients from 2004 to 2021 at a specialist centre: A longitudinal observational study. Eur J Surg Oncol. 2022;48:672-9. [PubMed: 34728141]

  • Chen JL, Miller DT, Schmidt LS, Malkin D, Korf BR, Eng C, Kwiatkowski DJ, Giannikou K. Mosaicism in tumor suppressor gene syndromes: prevalence, diagnostic strategies, and transmission risk. Annu Rev Genomics Hum Genet. 2022;23:331-61. [PubMed: 36044908]

  • Chen MY, Chew EY, Reynolds JC, Chao DL, Oldfield EH. Metastatic brainstem pheochromocytoma in a patient with von Hippel-Lindau disease. J Neurosurg. 2001;94:138. [PubMed: 11147885]

  • Chen X, Sanfilippo CJ, Nagiel A, Hosseini H, Mitchell D, McCannel CA, Schwartz SD, McCannel TA. Early detection of retinal hemangioblastomas in von Hippel-Lindau disease using ultra-widefield fluorescein angiography. Retina. 2018;38:748-754. [PubMed: 28368975]

  • Choo D, Shotland L, Mastroianni M, Glenn G, van Waes C, Linehan WM, Oldfield EH. Endolymphatic sac tumors in von Hippel-Lindau disease. J Neurosurg. 2004;100:480-7. [PubMed: 15035284]

  • Choueiri TK, Bauer TM, Papadopoulos KP, Plimack ER, Merchan JR, McDermott DF, Michaelson MD, Appleman LJ, Thamake S, Perini RF, Zojwalla NJ, Jonasch E. Inhibition of hypoxia-inducible factor-2α in renal cell carcinoma with belzutifan: a phase 1 trial and biomarker analysis. Nat Med. 2021;27:802-5. [PMC free article: PMC9128828] [PubMed: 33888901]

  • Coppin L, Grutzmacher C, Crépin M, Destailleur E, Giraud S, Cardot-Bauters C, Porchet N, Pigny P. VHL mosaicism can be detected by clinical next-generation sequencing and is not restricted to patients with a mild phenotype. Eur J Hum Genet. 2014;22:1149-52. [PMC free article: PMC4135403] [PubMed: 24301059]

  • Cybulski C, Krzystolik K, Murgia A, Gorski B, Debniak T, Jakubowska A, Martella M, Kurzawski G, Prost M, Kojder I, Limon J, Nowacki P, Sagan L, Bialas B, Kaluza J, Zdunek M, Omulecka A, Jaskolski D, Kostyk E, Koraszewska-Matuszewska B, Haus O, Janiszewska H, Pecold K, Starzycka M, Slomski R, Cwirko M, Sikorski A, Gliniewicz B, Cyrylowski L, Fiszer-Maliszewska L, Gronwald J, Toloczko-Grabarek A, Zajaczek S, Lubinski J. Germline mutations in the von Hippel-Lindau (VHL) gene in patients from Poland: disease presentation in patients with deletions of the entire VHL gene. J Med Genet. 2002;39:E38. [PMC free article: PMC1735187] [PubMed: 12114495]

  • Dalbah S, Bechrakis NE, Thomasen H, Flühs D, Rating P, Guberina M, Stuschke M, Fiorentzis M, Bornfeld N, Biewald E. Brachytherapy for peripheral retinal capillary haemangioblastoma in von Hippel-Lindau disease. Klin Monbl Augenheilkd. 2021;238:781-7. [PubMed: 34376008]

  • Dickson PV, Alex GC, Grubbs EG, Ayala-Ramirez M, Jimenez C, Evans DB, Lee JE, Perrier ND. Posterior retroperitoneoscopic adrenalectomy is a safe and effective alternative to transabdominal laparoscopic adrenalectomy for pheochromocytoma. Surgery. 2011;150:452-8. [PubMed: 21878230]

  • Dollfus H, Massin P, Taupin P, Nemeth C, Amara S, Giraud S, Beroud C, Dureau P, Gaudric A, Landais P, Richard S. Retinal hemangioblastoma in von Hippel-Lindau disease: a clinical and molecular study. Invest Ophthalmol Vis Sci. 2002;43:3067–74. [PubMed: 12202531]

  • Farhadi F, Nikpanah M, Li X, Symons R, Pourmorteza A, Merino MJ, Linehan WM, Malayeri AA. Germline VHL gene variant in patients with von Hippel-Lindau disease does not predict renal tumor growth. Abdom Radiol (NY). 2018;43:2743-9. [PMC free article: PMC8057112] [PubMed: 29525880]

  • Feletti A, Boaro A, Giampiccolo D, Casoli G, Moscolo F, Ferrara M, Sala F, Pavesi G. Spinal hemangioblastomas: analysis of surgical outcome and prognostic factors. Neurosurg Rev. 2022;45:1645-61. [PubMed: 34825301]

  • Frantzen C, Kruizinga RC, van Asselt SJ, Zonnenberg BA, Lenders JWM, de Herder WW, Walenkamp AME, Giles RH, Hes FJ, Sluiter WJ, van Pampus MG, Links TP. Pregnancy-related hemangioblastoma progression and complications in von Hippel-Lindau disease. Neurology. 2012;79:793-6. [PubMed: 22875085]

  • Friedman RA, Hoa M, Brackmann DE. Surgical management of endolymphatic sac tumors. J Neurol Surg B Skull Base. 2013;74:12-9. [PMC free article: PMC3699165] [PubMed: 24436884]

  • Gaudric A, Krivosic V, Duguid G, Massin P, Giraud S, Richard S. Vitreoretinal surgery for severe retinal capillary hemangiomas in von Hippel-Lindau disease. Ophthalmology. 2011;118:142-9. [PubMed: 20801520]

  • Giannini C, Scheithauer BW, Hellbusch LC, Rasmussen AG, Fox MW, McCormick SR, Davis DH. Peripheral nerve hemangioblastoma. Mod Pathol. 1998;11:999–1004. [PubMed: 9796730]

  • Giles RH, Gläsker S. The first prospective trial for von Hippel-Lindau disease: pazopanib. Lancet Oncol. 2018;19:1267-9. [PubMed: 30236512]

  • Gläsker S. Central nervous system manifestations in VHL: genetics, pathology and clinical phenotypic features. Fam Cancer. 2005;4:37-42. [PubMed: 15883708]

  • Gläsker S, Krüger MT, Klingler JH, Wlodarski M, Klompen J, Schatlo B, Hippchen B, Neumann HP, Van Velthoven V. Hemangioblastomas and neurogenic polyglobulia. Neurosurgery. 2013;72:930-5. [PubMed: 23407287]

  • Goyal N, Agrawal D, Singla R, Kale SS, Singh M, Sharma BS. Stereotactic radiosurgery in hemangioblastoma: experience over 14 years. J Neurosci Rural Pract. 2016;7:23-7. [PMC free article: PMC4750333] [PubMed: 26933339]

  • Groeben H, Walz MK, Nottebaum BJ, Alesina PF, Greenwald A, Schumann R, Hollmann MW, Schwarte L, Behrends M, Rössel T, Groeben C, Schäfer M, Lowery A, Hirata N, Yamakage M, Miller JA, Cherry TJ, Nelson A, Solorzano CC, Gigliotti B, Wang TS, Wietasch JKG, Friederich P, Sheppard B, Graham PH, Weingarten TN, Sprung J. International multicentre review of perioperative management and outcome for catecholamine-producing tumours. Br J Surg. 2020;107:e170-e178. [PMC free article: PMC8046358] [PubMed: 31903598]

  • Hes FJ, van der Luijt RB, Lips CJ. Clinical management of von Hippel–Lindau (VHL) disease. Neth J Med. 2001;59:225-34. [PubMed: 11705642]

  • Hrisomalos FN, Maturi RK, Pata V. Long-term use of intravitreal bevacizumab (avastin) for the treatment of von Hippel-Lindau associated retinal hemangioblastomas. Open Ophthalmol J 2010;4:66-9. [PMC free article: PMC3032222] [PubMed: 21293730]

  • Huang SJ, Amendola LM, Sternen DL. Variation among DNA banking consent forms: points for clinicians to bank on. J Community Genet. 2022;13:389-97. [PMC free article: PMC9314484] [PubMed: 35834113]

  • Huntoon K, Oldfield EH, Lonser RR. Dr. Arvid Lindau and discovery of von Hippel-Lindau disease. J Neurosurg. 2015;123:1093-7. [PubMed: 25748307]

  • Iliopoulos O. von Hippel-Lindau disease: genetic and clinical observations. Front Horm Res. 2001;28:131–66. [PubMed: 11443850]

  • Jimenez C, Cabanillas ME, Santarpia L, Jonasch E, Kyle KL, Lano EA, Matin SF, Nunez RF, Perrier ND, Phan A, Rich TA, Shah B, Williams MD, Waguespack SG. Use of the tyrosine kinase inhibitor sunitinib in a patient with von Hippel-Lindau disease: targeting angiogenic factors in pheochromocytoma and other von Hippel-Lindau disease-related tumors. J Clin Endocrinol Metab. 2009;94:386-91. [PubMed: 19017755]

  • Jonasch E, Donskov F, Iliopoulos O, Rathmell WK, Narayan VK, Maughan BL, Oudard S, Else T, Maranchie JK, Welsh SJ, Thamake S, Park EK, Perini RF, Linehan WM, Srinivasan R, et al. Belzutifan for renal cell carcinoma in von Hippel-Lindau disease. N Engl J Med. 2021;385:2036-46. [PMC free article: PMC9275515] [PubMed: 34818478]

  • Jonasch E, Iliopoulos O, Rathmell WK, Narayan V, Maughan BL, Oudard S, Else T, Maranchie JK, Welsh SJ, Iversen ABB, Roy A, Liu Y, Perini RF, Linehan WM, Srinivasan R. LITESPARK-004 (MK-6482-004) phase 2 study of belzutifan, an oral hypoxia-inducible factor 2α inhibitor (HIF-2α), for von Hippel-Lindau (VHL) disease: update with more than two years of follow-up data. J Clin Oncol. 2022;40:4546.

  • Jonasch E, McCutcheon IE, Gombos DS, Ahrar K, Perrier ND, Liu D, Robichaux CC, Villarreal MF, Weldon JA, Woodson AH, Pilie PG, Fuller GN, Waguespack SG, Matin SF. Pazopanib in patients with von Hippel-Lindau disease: a single-arm, single-centre, phase 2 trial. Lancet Oncol. 2018;19:1351-59. [PMC free article: PMC6338228] [PubMed: 30236511]

  • Jonasch E, McCutcheon IE, Waguespack SG, Wen S, Davis DW, Smith LA, Tannir NM, Gombos DS, Fuller GN, Matin SF. Pilot trial of sunitinib therapy in patients with von Hippel-Lindau disease. Ann Oncol. 2011;22:2661-6. [PMC free article: PMC4542805] [PubMed: 22105611]

  • Kano H, Shuto T, Iwai Y, Sheehan J, Yamamoto M, McBride HL, Sato M, Serizawa T, Yomo S, Moriki A, Kohda Y, Young B, Suzuki S, Kenai H, Duma C, Kikuchi Y, Mathieu D, Akabane A, Nagano O, Kondziolka D, Lunsford LD. Stereotactic radiosurgery for intracranial hemangioblastomas: a retrospective international outcome study. J Neurosurg. 2015;122:1469-78. [PubMed: 25816088]

  • Karimi S, Nikkhah H, Ahmadieh H, Safi S. Intravitreal injection of propranolol for the treatment of retinal capillary hemangioma in a case of von Hippel-Lindau. Retin Cases Brief Rep. 2020;14:305-9. [PubMed: 29466252]

  • Kim HJ, Butman JA, Brewer C, Zalewski C, Vortmeyer AO, Glenn G, Oldfield EH, Lonser RR. Tumors of the endolymphatic sac in patients with von Hippel-Lindau disease: implications for their natural history, diagnosis, and treatment. J Neurosurg. 2005; 102:503-12. [PubMed: 15796386]

  • Kim HJ, Hagan M, Butman JA, Baggenstos M, Brewer C, Zalewski C, Linehan WM, Lonser RR. Surgical resection of endolymphatic sac tumors in von Hippel-Lindau disease: findings, results, and indications. Laryngoscope. 2013;123:477-83. [PMC free article: PMC7560989] [PubMed: 23070752]

  • Kim WY, Kaelin WG. Role of VHL gene mutations in human cancer. J Clin Oncol. 2004;22:4991-5004. [PubMed: 15611513]

  • Knauth K, Bex C, Jemth P, Buchberger A. Renal cell carcinoma risk in type 2 von Hippel-Lindau disease correlates with defects in pVHL stability and HIF-1alpha interactions. Oncogene. 2006;25:370–7. [PubMed: 16261165]

  • Krauss T, Ferrara AM, Links TP, Wellner U, Bancos I, Kvachenyuk A, Villar Gómez de Las Heras K, Yukina MY, Petrov R, Bullivant G, von Duecker L, Jadhav S, Ploeckinger U, Welin S, Schalin-Jäntti C, Gimm O, Pfeifer M, Ngeow J, Hasse-Lazar K, Sansó G, Qi X, Ugurlu MU, Diaz RE, Wohllk N, Peczkowska M, Aberle J, Lourenço DM Jr, Pereira MAA, Fragoso MCBV, Hoff AO, Almeida MQ, Violante AHD, Quidute ARP, Zhang Z, Recasens M, Díaz LR, Kunavisarut T, Wannachalee T, Sirinvaravong S, Jonasch E, Grozinsky-Glasberg S, Fraenkel M, Beltsevich D, Egorov VI, Bausch D, Schott M, Tiling N, Pennelli G, Zschiedrich S, Därr R, Ruf J, Denecke T, Link KH, Zovato S, von Dobschuetz E, Yaremchuk S, Amthauer H, Makay Ö, Patocs A, Walz MK, Huber TB, Seufert J, Hellman P, Kim RH, Kuchinskaya E, Schiavi F, Malinoc A, Reisch N, Jarzab B, Barontini M, Januszewicz A, Shah N, Young WF Jr, Opocher G, Eng C, Neumann HPH, Bausch B. Preventive medicine of von Hippel-Lindau disease-associated pancreatic neuroendocrine tumors. Endocr Relat Cancer. 2018;25:783-93. [PubMed: 29748190]

  • Kreusel KM. Ophthalmological manifestations in VHL and NF 1: pathological and diagnostic implications. Fam Cancer. 2005;4:43–7. [PubMed: 15883709]

  • Kreusel KM, Bechrakis NE, Krause L, Neumann HP, Foerster MH. Retinal angiomatosis in von Hippel-Lindau disease: a longitudinal ophthalmologic study. Ophthalmology. 2006;113:1418–24. [PubMed: 16769118]

  • Krüger MT, Steiert C, Gläsker S, Klingler JH. Minimally invasive resection of spinal hemangioblastoma: feasibility and clinical results in a series of 18 patients. J Neurosurg Spine. 2019;31:880-89. [PubMed: 31398701]

  • Kwon T, Jeong IG, Pak S, You D, Song C, Hong JH, Ahn H, Kim CS. Renal tumor size is an independent prognostic factor for overall survival in von Hippel-Lindau disease. J Cancer Res Clin Oncol. 2014;140:1171-7. [PubMed: 24671227]

  • Lammens CR, Aaronson NK, Hes FJ, Links TP, Zonnenberg BA, Lenders JW, Majoor-Krakauer D, Van Os TA, Gomez-Garcia EB, de Herder W, van der Luijt RB, van den Ouweland AM, Van Hest LP, Verhoef S, Bleiker EM. Compliance with period surveillance for von Hippel-Lindau disease. Genet Med. 2011a;13:519-27. [PubMed: 21415761]

  • Lammens CR, Bleiker EM, Verhoef S, Ausems MG, Majoor-Krakauer D, Sijmons RH, Hes FJ, Gómez-García EB, Van Os TA, Spruijt L, van der Luijt RB, van den Ouweland AM, Ruijs MW, Gundy C, Nagtegaal T, Aaronson NK. Distress in partners of individuals diagnosed with or at high risk of developing tumors due to rare hereditary cancer syndromes. Psychooncology. 2011b;20:631-8. [PubMed: 21384469]

  • Lammens CR, Bleiker EM, Verhoef S, Hes FJ, Ausems MG, Majoor-Krakauer D, Sijmons RH, van der Luijt RB, van den Ouweland AM, Van Os TA, Hoogerbrugge N, Gómez García EB, Dommering CJ, Gundy CM, Aaronson NK. Psychosocial impact of von Hippel-Lindau disease: levels and sources of distress. Clin Genet. 2010;77:483-91. [PubMed: 20184621]

  • Lenglet M, Robriquet F, Schwarz K, Camps C, Couturier A, Hoogewijs D, Buffet A, Knight SJL, Gad S, Couvé S, Chesnel F, Pacault M, Lindenbaum P, Job S, Dumont S, Besnard T, Cornec M, Dreau H, Pentony M, Kvikstad E, Deveaux S, Burnichon N, Ferlicot S, Vilaine M, Mazzella JM, Airaud F, Garrec C, Heidet L, Irtan S, Mantadakis E, Bouchireb K, Debatin KM, Redon R, Bezieau S, Bressac-de Paillerets B, Teh BT, Girodon F, Randi ML, Putti MC, Bours V, Van Wijk R, Göthert JR, Kattamis A, Janin N, Bento C, Taylor JC, Arlot-Bonnemains Y, Richard S, Gimenez-Roqueplo AP, Cario H, Gardie B. Identification of a new VHL exon and complex splicing alterations in familial erythrocytosis or von Hippel-Lindau disease. Blood. 2018;132:469-83. [PubMed: 29891534]

  • Li SR, Nicholson KJ, Mccoy KL, Carty SE, Yip L. Clinical and biochemical features of pheochromocytoma characteristic of von Hippel-Lindau Syndrome. World J Surg. 2020;44:570-77. [PubMed: 31773218]

  • Lonser RR, Butman JA, Huntoon K, Asthagiri AR, Wu T, Bakhtian KD, Chew EY, Zhuang Z, Linehan WM, Oldfield EH. Prospective natural history study of central nervous system hemangioblastomas in von Hippel-Lindau disease. J Neurosurg. 2014;120:1055-62. [PMC free article: PMC4762041] [PubMed: 24579662]

  • Lonser RR, Butman JA, Kiringoda R, Song D, Oldfield EH. Pituitary stalk hemangioblastomas in von Hippel-Lindau disease. J Neurosurg. 2009;110:350-3. [PMC free article: PMC2770699] [PubMed: 18834262]

  • Lonser RR, Glenn GM, Walther M, Chew EY, Libutti SK, Linehan WM, Oldfield EH. von Hippel-Lindau disease. Lancet. 2003;361:2059-67. [PubMed: 12814730]

  • Madhusudan S, Deplanque G, Braybrooke JP, Cattell E, Taylor M, Price P, Tsaloumas MD, Moore N, Huson SM, Adams C, Frith P, Scigalla P, Harris AL. Antiangiogenic therapy for von Hippel-Lindau disease. JAMA. 2004;291:943-4. [PubMed: 14982909]

  • Maher ER, Iselius L, Yates JR, Littler M, Benjamin C, Harris R, Sampson J, Williams A, Ferguson-Smith MA, Morton N. Von Hippel-Lindau disease: a genetic study. J Med Genet. 1991;28:443–7. [PMC free article: PMC1016952] [PubMed: 1895313]

  • Maher ER, Neumann HP, Richard S. von Hippel-Lindau disease: a clinical and scientific review. Eur J Hum Genet. 2011;19:617-23. [PMC free article: PMC3110036] [PubMed: 21386872]

  • Maher ER, Yates RJ, Harries R, Benjamin R, Moorre AT, Ferguson-Smith MA. Clinical features and natural history of von Hippel-Lindau disease. Q J Med. 1990;77:1151-63. [PubMed: 2274658]

  • Maranchie JK, Afonso A, Albert PS, Kalyandrug S, Phillips JL, Zhou S, Peterson J, Ghadimi BM, Hurley K, Riss J, Vasselli JR, Ried T, Zbar B, Choyke P, Walther MM, Klausner RD, Linehan WM. Solid renal tumor severity in von Hippel Lindau disease is related to germline deletion length and location. Hum Mutat. 2004;23:40-6. [PubMed: 14695531]

  • Mazzapicchi E, Restelli F, Falco J, Broggi M, Gatti L, Alongi P, Valentini L, Ferroli P, Vetrano IG, DiMeco F, Acerbi F. Sporadic and von Hippel-Lindau related hemangioblastomas of brain and spinal cord: multimodal imaging for intraoperative strategy. Cancers (Basel). 2022;14:5492. [PMC free article: PMC9688688] [PubMed: 36428588]

  • McNeill A, Rattenberry E, Barber R, Killick P, MacDonald F, Maher ER. Genotype-phenotype correlations in VHL exon deletions. Am J Med Genet A. 2009;149A:2147-51. [PubMed: 19764026]

  • Mehta GU, Kim HJ, Gidley PW, Daniels AB, Miller ME, Lekovic GP, Butman JA, Lonser RR. Endolymphatic sac tumor screening and diagnosis in von Hippel-Lindau disease: a consensus statement. J Neurol Surg B Skull Base. 2021;83:e225-e231. [PMC free article: PMC9272323] [PubMed: 35833006]

  • Mehta GU, Montgomery BK, Maggio DM, Chittiboina P, Oldfield EH, Lonser RR. Functional outcome after resection of von Hippel-Lindau disease-associated cauda equina hemangioblastomas: an observational cohort study. Oper Neurosurg (Hagerstown). 2017;13:435-40. [PMC free article: PMC6280992] [PubMed: 28838111]

  • Metelo AM, Noonan H, Iliopoulos O. HIF2a inhibitors for the treatment of VHL disease. Oncotarget. 2015;6:23036-7. [PMC free article: PMC4695098] [PubMed: 26325097]

  • Migliorini D, Haller S, Merkler D, Pugliesi-Rinaldi A, Koka A, Schaller K, Leemann B, Dietrich PY. Recurrent multiple CNS hemangioblastomas with VHL disease treated with pazopanib: a case report and literature review. CNS Oncol. 2015;4:387-92. [PMC free article: PMC6083944] [PubMed: 26497655]

  • Molino D, Sepe J, Anastasio P, De Santo NG. The history of von Hippel-Lindau disease. J Nephrol. 2006;19:S119-23. [PubMed: 16874724]

  • Murgia A, Martella M, Vinanzi C, Polli R, Perilongo G, Opocher G. Somatic mosaicism in von Hippel-Lindau Disease. Hum Mutat. 2000;15:114. [PubMed: 10612832]

  • Muzumdar DP, Goel A, Fattepurkar S, Goel N. Endolymphatic sac carcinoma of the right petrous bone in Von Hippel-Lindau disease. J Clin Neurosci. 2006;13:471–4. [PubMed: 16678727]

  • Nambu S, Otani R, Higuchi F, Uzuka T, Matsuda H, Kim P, Ueki K. Histology of hemangioblastoma treated with stereotactic radiosurgery confirms its effectiveness. J Clin Neurosci. 2018;51:43-5. [PubMed: 29483011]

  • Neumann HP, Eng C. The approach to the patient with paraganglioma. J Clin Endocrinol Metab. 2009;94:2677-83. [PMC free article: PMC2730863] [PubMed: 19657044]

  • Neumann HPH, Tsoy U, Bancos I, Amodru V, Walz MK, Tirosh A, Kaur RJ, McKenzie T, Qi X, Bandgar T, Petrov R, Yukina MY, Roslyakova A, van der Horst-Schrivers ANA, Berends AMA, Hoff AO, Castroneves LA, Ferrara AM, Rizzati S, Mian C, Dvorakova S, Hasse-Lazar K, Kvachenyuk A, Peczkowska M, Loli P, Erenler F, Krauss T, Almeida MQ, Liu L, Zhu F, Recasens M, Wohllk N, Corssmit EPM, Shafigullina Z, Calissendorff J, Grozinsky-Glasberg S, Kunavisarut T, Schalin-Jäntti C, Castinetti F, Vlcek P, Beltsevich D, Egorov VI, Schiavi F, Links TP, Lechan RM, Bausch B, Young WF Jr, Eng C, et al. comparison of pheochromocytoma-specific morbidity and mortality among adults with bilateral pheochromocytomas undergoing total adrenalectomy vs cortical-sparing adrenalectomy. JAMA Netw Open. 2019;2:e198898. [PMC free article: PMC6692838] [PubMed: 31397861]

  • Nevoux J, Nowak C, Vellin JF, Lepajolec C, Sterkers O, Richard S, Bobin S. Management of endolymphatic sac tumors: sporadic cases and von Hippel-Lindau disease. Otol Neurotol. 2014;35:899-904. [PubMed: 24662627]

  • Nordstrom-O'Brien M, van der Luijt RB, van Rooijen E, van den Ouweland AM, Majoor-Krakauer DF, Lolkema MP, van Brussel A, Voest EE, Giles RH. Genetic analysis of von Hippel-Lindau disease. Hum Mutat. 2010;31:521-37. [PubMed: 20151405]

  • Oosting SF, van Asselt SJ, Brouwers AH, Bongaerts AH, Steinberg JD, de Jong JR, Lub-de Hooge MN, van der Horst-Schrivers AN, Walenkamp AM, Hoving EW, Sluiter WJ, Zonnenberg BA, de Vries EG, Links TP. 89Zr-bevacizumab PET visualizes disease manifestations in patients with von Hippel-Lindau disease. J Nucl Med. 2016;57:1244-50. [PubMed: 27173161]

  • O'Toole SM, Sahdev A, Bhattacharya S, Feakins R, Gevers EF, Drake WM. Paediatric pancreatic neuroendocrine tumours in von Hippel-Lindau disease. Endocr Relat Cancer. 2018;25:L43-7. [PubMed: 29752349]

  • Pan J, Ho AL, D'Astous M, Sussman ES, Thompson PA, Tayag AT, Pangilinan L, Soltys SG, Gibbs IC, Chang SD. Image-guided stereotactic radiosurgery for treatment of spinal hemangioblastoma. Neurosurg Focus. 2017;42:E12. [PubMed: 28041328]

  • Pan J, Jabarkheel R, Huang Y, Ho A, Chang SD. Stereotactic radiosurgery for central nervous system hemangioblastoma: systematic review and meta-analysis. J Neurooncol. 2018;137:11-22. [PubMed: 29204841]

  • Peng X, Chen J, Wang J, Peng S, Liu S, Ma K, Zhou J, Hong B, Zhou B, Zhang J, Cai L, Gong K. Natural history of renal tumours in von Hippel-Lindau disease: a large retrospective study of Chinese patients. J Med Genet. 2019;56:380-7. [PubMed: 30745424]

  • Pilié P, Hasanov E, Matin SF, Woodson AHH, Marcott VD, Bird S, Slack RS, Fuller GN, McCutcheon IE, Jonasch E. Pilot study of dovitinib in patients with von Hippel-Lindau disease. Oncotarget. 2018;9:23390-5. [PMC free article: PMC5955100] [PubMed: 29805741]

  • Priesemann M, Davies KM, Perry LA, Drake WM, Chew SL, Monson JP, Savage MO, Johnston LB. Benefits of screening in von Hippel-Lindau disease--comparison of morbidity associated with initial tumours in affected parents and children. Horm Res. 2006;66:1-5. [PubMed: 16651847]

  • Raja D, Benz MS, Murray TG, Escalona-Benz EM, Markoe A. Salvage external beam radiotherapy of retinal capillary hemangiomas secondary to von Hippel-Lindau disease: visual and anatomic outcomes. Ophthalmology. 2004;111:150-3. [PubMed: 14711727]

  • Reich M, Jaegle S, Neumann-Haefelin E, Klingler JH, Evers C, Daniel M, Bucher F, Ludwig F, Nuessle S, Kopp J, Boehringer D, Reinhard T, Lagrèze WA, Lange C, Agostini H, Lang SJ. Genotype-phenotype correlation in von Hippel-Lindau disease. Acta Ophthalmol. 2021;99:e1492-e1500. [PubMed: 33720516]

  • Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, Grody WW, Hegde M, Lyon E, Spector E, Voelkerding K, Rehm HL, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17:405-24. [PMC free article: PMC4544753] [PubMed: 25741868]

  • Robson ME, Bradbury AR, Arun B, Domchek SM, Ford JM, Hampel HL, Lipkin SM, Syngal S, Wollins DS, Lindor NM. American Society of Clinical Oncology policy statement update: genetic and genomic testing for cancer susceptibility. J Clin Oncol. 2015;33:3660-7. [PubMed: 26324357]

  • Sanford T, Gomella PT, Siddiqui R, Su D, An JY, Bratslavsky G, Ball MW, Linehan WM, Metwalli AR. Long term outcomes for patients with von Hippel-Lindau and Pheochromocytoma: defining the role of active surveillance. Urol Oncol. 2021;39:134.e1-134.e8. [PMC free article: PMC9175510] [PubMed: 33303379]

  • Sansó G, Rudaz MC, Levin G, Barontini M. Familial isolated pheochromocytoma presenting a new mutation in the von Hippel-Lindau gene. Am J Hypertens. 2004;17:1107-11. [PubMed: 15607616]

  • Santarpia L, Sarlis NJ, Santarpia M, Sherman SI, Trimarchi F, Benvenga S. Mosaicism in von Hippel-Lindau disease: an event important to recognize. J Cell Mol Med. 2007;11:1408-15. [PMC free article: PMC4401302] [PubMed: 18205710]

  • Sardi I, Sanzo M, Giordano F, Buccoliero AM, Mussa F, Aricò M, Genitori L. Monotherapy with thalidomide for treatment of spinal cord hemangioblastomas in a patient with von Hippel-Lindau disease. Pediatr Blood Cancer. 2009;53:464-7. [PubMed: 19415739]

  • Schimke RN, Collins DL, Rothberg PG. Functioning carotid paraganglioma in the von Hippel-Lindau syndrome [letter]. Am J Med Genet. 1998;80:533-4. [PubMed: 9880225]

  • Sgambati MT, Stolle C, Choyke PL, Walther MM, Zbar B, Linehan WM, Glenn GM. Mosaicism in von Hippel-Lindau disease: lessons from kindreds with germline mutations identified in offspring with mosaic parents. Am J Hum Genet. 2000;66:84-91. [PMC free article: PMC1288351] [PubMed: 10631138]

  • Sharma A, Mukewar S, Vege SS. Clinical profile of pancreatic cystic lesions in von Hippel-Lindau disease: a series of 48 patients seen at a tertiary institution. Pancreas. 2017;46:948-52. [PubMed: 28697137]

  • Shirali AS, Clemente-Gutierrez U, Huang BL, Lui MS, Chiang YJ, Jimenez C, Fisher SB, Graham PH, Lee JE, Grubbs EG, Perrier ND. Pheochromocytoma recurrence in hereditary disease: does a cortical-sparing technique increase recurrence rate? Surgery. 2023;173:26-34. [PubMed: 36229248]

  • Siller S, Szelényi A, Herlitz L, Tonn JC, Zausinger S. Spinal cord hemangioblastomas: significance of intraoperative neurophysiological monitoring for resection and long-term outcome. J Neurosurg Spine. 2017;26:483-93. [PubMed: 27982764]

  • Simone CB, Lonser RR, Ondos J, Oldfield EH, Camphausen K, Simone NL. Infratentorial craniospinal irradiation for von Hippel-Lindau: a retrospective study supporting a new treatment for patients with CNS hemangioblastomas. Neuro Oncol. 2011;13:1030-6. [PMC free article: PMC3158017] [PubMed: 21798886]

  • Sizdahkhani S, Feldman MJ, Piazza MG, Ksendzovsky A, Edwards NA, Ray-Chaudhury A, Maric D, Merrill MJ, Pacak K, Zhuang Z, Chittiboina P. Somatostatin receptor expression on von Hippel-Lindau-associated hemangioblastomas offers novel therapeutic target. Sci Rep. 2017;7:40822. [PMC free article: PMC5240113] [PubMed: 28094316]

  • Stebbins CE, Kaelin WG Jr, Pavletich NP. Structure of the VHL-ElonginC-ElonginB complex: implications for VHL tumor suppressor function. Science. 1999;284:455–61. [PubMed: 10205047]

  • Stenson PD, Mort M, Ball EV, Chapman M, Evans K, Azevedo L, Hayden M, Heywood S, Millar DS, Phillips AD, Cooper DN. The Human Gene Mutation Database (HGMD®): optimizing its use in a clinical diagnostic or research setting. Hum Genet. 2020;139:1197-207. [PMC free article: PMC7497289] [PubMed: 32596782]

  • Takami H, Graffeo CS, Perry A, Brown DA, Meyer FB, Burns TC, Parney IF. Presentation, imaging, patterns of care, growth, and outcome in sporadic and von Hippel-Lindau-associated central nervous system hemangioblastomas. J Neurooncol. 2022;159:221-31. [PubMed: 35902552]

  • Takamori H, Yamasaki T, Kitadai R, Minamishima YA, Nakamura E. Development of drugs targeting hypoxia-inducible factor against tumor cells with VHL mutation: story of 127 years. Cancer Sci. 2023;114:1208-17. [PMC free article: PMC10067407] [PubMed: 36650918]

  • Tirosh A, El Lakis M, Green P, Nockel P, Patel D, Nilubol N, Gara SK, Keutgen XM, Linehan WM, Kebebew E. In silico VHL gene mutation analysis and prognosis of pancreatic neuroendocrine tumors in von Hippel-Lindau disease. J Clin Endocrinol Metab. 2018a;103:1631-8. [PMC free article: PMC6276699] [PubMed: 29294023]

  • Tirosh A, Sadowski SM, Linehan WM, Libutti SK, Patel D, Nilubol N, Kebebew E. Association of VHL genotype with pancreatic neuroendocrine tumor phenotype in patients with von Hippel-Lindau disease. JAMA Oncol. 2018b;4:124-6. [PMC free article: PMC5833646] [PubMed: 29075773]

  • van Overdam KA, Missotten T, Kilic E, Spielberg LH. Early surgical treatment of retinal hemangioblastomas. Acta Ophthalmol. 2017;95:97-102. [PubMed: 27681600]

  • Volkin D, Yerram N, Ahmed F, Lankford D, Baccala A, Gupta GN, Hoang A, Nix J, Metwalli AR, Lang DM, Bratslavsky G, Linehan WM, Pinto PA. Partial adrenalectomy minimizes the need for long-term hormone replacement in pediatric patients with pheochromocytoma and von Hippel-Lindau syndrome. J Pediatr Surg. 2012;47:2077-82. [PMC free article: PMC3846393] [PubMed: 23164001]

  • Wanebo JE, Lonser RR, Glenn GM, Oldfield EH. The natural history of hemangioblastomas of the central nervous system in patients with von Hippel-Lindau disease. J Neurosurg. 2003;98:82–94. [PubMed: 12546356]

  • Webster AR, Maher ER, Moore AT. Clinical characteristics of ocular angiomatosis in von Hippel-Lindau disease and correlation with germline mutation. Arch Ophthalmol. 1999;117:371–8. [PubMed: 10088816]

  • Weirich G, Klein B, Wohl T, Engelhardt D, Brauch H. VHL2C phenotype in a German von Hippel-Lindau family with concurrent VHL germline mutations P81S and L188V. J Clin Endocrinol Metab. 2002;87:5241–6. [PubMed: 12414898]

  • Wessendorf J, König A, Heers H, Mahnken AH. Repeat percutaneous radiofrequency ablation of T1 renal cell carcinomas is safe in patients with von Hippel-Lindau disease. Cardiovasc Intervent Radiol. 2021;44:2022-5. [PMC free article: PMC8626382] [PubMed: 34414496]

  • Wilding A, Ingham SL, Lalloo F, Clancy T, Huson SM, Moran A, Evans DG. Life expectancy in hereditary cancer predisposing diseases: an observational study. J Med Genet. 2012;49:264-9. [PubMed: 22362873]

  • Wilschanski M, Miller LL, Shoseyov D, Blau H, Rivlin J, Aviram M, Cohen M, Armoni S, Yaakov Y, Pugatsch T, Cohen-Cymberknoh M, Miller NL, Reha A, Northcutt VJ, Hirawat S, Donnelly K, Elfring GL, Ajayi T, Kerem E. Chronic ataluren (PTC124) treatment of nonsense mutation cystic fibrosis. Eur Respir J. 2011;38:59-69. [PubMed: 21233271]

  • Wolters WPG, Dreijerink KMA, Giles RH, van der Horst-Schrivers ANA, van Nesselrooij B, Zandee WT, Timmers HJLM, Seute T, de Herder WW, Verrijn Stuart AA, Kilic E, Brinkman WM, Zondervan PJ, Vandertop WP, Daniels AB, Wolbers T, Links TP, van Leeuwaarde RS. Multidisciplinary integrated care pathway for von Hippel-Lindau disease. Cancer. 2022;128:2871-79. [PMC free article: PMC9542729] [PubMed: 35579632]

  • Wong WT, Liang KJ, Hammel K, Coleman HR, Chew EY. Intravitreal ranibizumab therapy for retinal capillary hemangioblastoma related to von Hippel-Lindau disease. Ophthalmology. 2008;115:1957-64. [PMC free article: PMC3034164] [PubMed: 18789534]

  • Wu P, Zhang N, Wang X, Li T, Ning X, Bu D, Gong K. Mosaicism in von Hippel-Lindau disease with severe renal manifestations. Clin Genet. 2013;84:581-4. [PubMed: 23384228]

  • Ye DY, Bakhtian KD, Asthagiri AR, Lonser RR. Effect of pregnancy on hemangioblastoma development and progression in von Hippel-Lindau disease. J Neurosurg. 2012;117:818-24. [PubMed: 22937928]

  • Young EE, Castle SM, Gorbatiy V, Leveillee RJ. Comparison of safety, renal function outcomes and efficacy of laparoscopic and percutaneous radio frequency ablation of renal masses. J Urol. 2012;187:1177-82. [PubMed: 22357170]