U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Adam MP, Feldman J, Mirzaa GM, et al., editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993-2024.

Cover of GeneReviews®

GeneReviews® [Internet].

Show details

Noonan Syndrome

, MD.

Author Information and Affiliations

Initial Posting: ; Last Revision: February 17, 2022.

Estimated reading time: 1 hour

Summary

Clinical characteristics.

Noonan syndrome (NS) is characterized by characteristic facies, short stature, congenital heart defect, and developmental delay of variable degree. Other findings can include broad or webbed neck, unusual chest shape with superior pectus carinatum and inferior pectus excavatum, cryptorchidism, varied coagulation defects, lymphatic dysplasias, and ocular abnormalities. Although birth length is usually normal, final adult height approaches the lower limit of normal. Congenital heart disease occurs in 50%-80% of individuals. Pulmonary valve stenosis, often with dysplasia, is the most common heart defect and is found in 20%-50% of individuals. Hypertrophic cardiomyopathy, found in 20%-30% of individuals, may be present at birth or develop in infancy or childhood. Other structural defects include atrial and ventricular septal defects, branch pulmonary artery stenosis, and tetralogy of Fallot. Up to one fourth of affected individuals have mild intellectual disability, and language impairments in general are more common in NS than in the general population.

Diagnosis/testing.

The diagnosis of Noonan is established in a proband with suggestive findings and a heterozygous pathogenic variant in BRAF, KRAS, MAP2K1, MRAS, NRAS, PTPN11, RAF1, RASA2, RIT1, RRAS2, SOS1, or SOS2 or either a heterozygous variant or biallelic pathogenic variants in LZTR1 identified by molecular genetic testing. Several additional genes associated with a Noonan syndrome-like phenotype in fewer than ten individuals have been identified.

Management.

Treatment of manifestations: Cardiovascular anomalies in NS are usually treated as in the general population. Developmental disabilities are addressed by early intervention programs and individualized education strategies. Treatment for serious bleeding is guided by knowledge of the specific factor deficiency or platelet aggregation anomaly. Growth hormone (GH) treatment increases growth velocity. Standard treatment for juvenile myelomonocytic leukemia (JMML) and other malignancies, feeding difficulties, ADHD, behavioral problems, cryptorchidism in males, renal anomalies / hydronephrosis, strabismus, hearing loss, and Chiari malformation.

Surveillance: At each visit: measurement of growth parameters; evaluation of nutritional status in infants and toddlers; monitor for evidence of new neurologic manifestations (chronic headache, neck pain, changes in tone, dizziness, or obstructive sleep apnea); monitor developmental progress; assessment of behavioral issues, as age appropriate; skin examination. Annually in childhood or as clinically indicated: ophthalmology and audiology evaluations.

  • In children age <5 years: if initial cardiac evaluation is normal, at least annual cardiac evaluations until age 5 years.
  • In children age >5 years through adulthood, cardiac evaluation at least every 5 years, or as clinically indicated.
  • Prior to any surgical procedure or in those with clinical bleeding: assessment of bleeding history, CBC with differential, and consideration of measurement of coagulation factors.
  • For those with pathogenic PTPN11 or KRAS variants: consider physical examination with assessment of spleen size & CBC every 3-6 months until age 5 years to assess for concerns about JMML/malignancy.

Agents/circumstances to avoid: Aspirin therapy should be avoided because it may exacerbate a bleeding diathesis.

Pregnancy management: Consider referral to an adult congenital heart program for peripartum evaluation and management; consider a hematology referral if the affected pregnant woman has a history of bleeding abnormalities and/or has not undergone previous screening for coagulopathy.

Genetic counseling.

NS is most often inherited in an autosomal dominant manner. While many individuals with autosomal dominant NS have a de novo pathogenic variant, an affected parent is recognized in 30%-75% of families. The risk to sibs of a proband with autosomal dominant NS depends on the genetic status of the parents: if a parent is affected, the risk is 50%; when the parents are clinically unaffected, the risk to the sibs of a proband appears to be low (<1%). Each child of an individual with autosomal dominant NS has a 50% chance of inheriting the pathogenic variant.

NS caused by pathogenic variants in LZTR1 can be inherited in either an autosomal dominant or an autosomal recessive manner. The parents of an individual with autosomal recessive NS are typically heterozygotes (i.e., have one LZTR1 pathogenic variant), and may either be asymptomatic or have mild features of NS. If both parents are heterozygous for one LZTR1 pathogenic variant, each sib of an affected individual has at conception a 25% chance of being affected, a 50% chance of having one LZTR1 pathogenic variant (which can be associated with mild NS features), and a 25% chance of being unaffected and not a carrier.

Prenatal testing and preimplantation genetic testing are possible if the NS-related pathogenic variant(s) have been identified in an affected family member.

Diagnosis

No consensus clinical diagnostic criteria for Noonan syndrome have been published. Diagnostic scoring systems, most recently published in van der Burgt [2007] and embedded in the management guidelines developed by DYSCERNE in the United Kingdom [Noonan Syndrome Guideline Development Group 2010], have been proposed but have not been used extensively in North America.

Suggestive Findings

Noonan syndrome (NS) should be suspected in individuals with the following clinical, laboratory, and family history findings.

Clinical findings

  • Characteristic facies. The facial appearance of NS shows considerable change with age, being most striking in young and middle childhood, and most subtle in adulthood. Key features found regardless of age include the following:
    • Low-set, posteriorly rotated ears with fleshy helices
    • Vivid blue or blue-green irises
    • Widely spaced and downslanted palpebral fissures
    • Epicanthal folds
    • Fullness or droopiness of the upper eyelids (ptosis)
    Note: See the National Human Genome Research Institute (NHGRI) Atlas of Human Malformation Syndromes (scroll to ATLAS IMAGES) for photographs of individuals with Noonan syndrome from diverse ethnic backgrounds.
  • Short stature for sex and family background
  • Congenital heart defects, most commonly pulmonary valve stenosis, atrial septal defect, and/or hypertrophic cardiomyopathy
  • Developmental delay of variable degree
  • Broad or webbed neck
  • Unusual chest shape with superior pectus carinatum and inferior pectus excavatum
  • Widely spaced nipples
  • Cryptorchidism in males
  • Lymphatic dysplasia of the lungs, intestines, and/or lower extremities

Suggestive laboratory findings. Coagulation defects:

  • Coagulation screens (e.g., prothrombin time, activated partial thromboplastin time, platelet count, and platelet aggregation testing) may show abnormalities.
  • Specific testing should identify the particular coagulation defect, such as von Willebrand disease, thrombocytopenia, varied coagulation factor defects (factors V, VIII, XI, XII, protein C), and platelet dysfunction.

Family history is consistent with autosomal dominant (e.g., affected males and females in multiple generations) or ‒ rarely ‒ autosomal recessive inheritance (see Genetic Counseling). Absence of a known family history of Noonan syndrome does not preclude the diagnosis.

Establishing the Diagnosis

The molecular diagnosis of NS is established in a proband with suggestive findings and a heterozygous pathogenic (or likely pathogenic) variant in one of the genes listed in Table 1 or biallelic pathogenic (or likely pathogenic) variants in LZTR1.

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 section is understood to include likely pathogenic variants. (2) The identification of variant(s) of uncertain significance cannot be used to confirm or rule out the diagnosis. (3) As up to 20% of individuals meeting the proposed clinical diagnostic criteria [van der Burgt 2007] for NS do not have an identifiable molecular genetic etiology, nondiagnostic genetic testing does not rule out the diagnosis.

Molecular genetic testing approaches can include a combination of gene-targeted testing (multigene panel) and comprehensive genomic testing (exome sequencing or genome sequencing) depending on the phenotype.

Gene-targeted testing requires that the clinician determine which gene(s) are likely involved, whereas genomic testing does not. Individuals with the distinctive findings described in Suggestive Findings are likely to be diagnosed using gene-targeted testing (see Option 1), whereas those in whom the diagnosis of Noonan syndrome has not been considered are more likely to be diagnosed using genomic testing (see Option 2).

Option 1

When the phenotypic findings suggest the diagnosis of Noonan syndrome, molecular genetic testing approaches usually include the use of a multigene panel. A Noonan syndrome multigene panel that includes some or all of the genes listed in Table 1 and other genes of interest (see Differential Diagnosis) is most likely 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. Since Noonan syndrome occurs through a gain-of-function mechanism and large intragenic deletions or duplications have not been reported, testing for intragenic deletions or duplications is unlikely to result in a diagnosis; however, rare cases have been reported for some genes (see Table 1).

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

Note: Serial single-gene testing can be considered if panel testing is not feasible. Approximately 50% of individuals with NS have a pathogenic missense variant in PTPN11; therefore, single-gene testing starting with PTPN11 would be the next best first test. Appropriate serial single-gene testing if PTPN11 testing is not diagnostic can be determined by the individual's phenotype (e.g., RIT1 if there is hypertrophic cardiomyopathy, LZTR1 if autosomal recessive inheritance is suspected); however, continued sequential single-gene testing is not recommended as it is less efficient and more costly than panel testing.

Option 2

When the diagnosis of Noonan syndrome has not been considered because an individual has atypical phenotypic features or if some but not all characteristic phenotypic features are present (e.g., a "Noonan-like" phenotype), comprehensive genomic testing, which does not require the clinical to determine which gene is likely involved, may be used. 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 1.

Molecular Genetic Testing Used in Noonan Syndrome (NS)

Gene 1, 2Proportion of NS Attributed to Pathogenic Variants in GeneProportion of Probands with a Pathogenic Variant 3 Detected by Method
Sequence analysis 4Gene-targeted deletion/duplication analysis 5
BRAF <2% 6100%Unknown 7
KRAS <5% 8100%Unknown 7
LZTR1 ~8% 9100%Unknown 7
MAP2K1 <2% 10100%Unknown 7
MRAS <1% 11100%Unknown 7
NRAS <1% 12100%Unknown 7
PTPN11 50% 13Nearly 100%Rare duplication14 diagnosis of NS questioned 15
RAF1 5% 16Nearly 100%1 reported case w/a duplication17 diagnosis of NS questioned 15; 1 reported case of a deletion 18
RASA2 Unknown 19100%Unknown 7
RIT1 5% 16100%Unknown 7
RRAS2 <1% 20100%Unknown 7
SOS1 10%-13% 21100%Unknown 7
SOS2 ~4% 22100%Unknown 7
Others 23NA
1.

Genes are listed in alphabetic order.

2.
3.

See Molecular Genetics for information on variants detected in these genes

4.

Sequence analysis detects variants that are benign, likely benign, of uncertain significance, likely pathogenic, or pathogenic. Variants may include small intragenic deletions/insertions and missense, nonsense, and splice site variants; typically, exon or whole-gene deletions/duplications are not detected. For issues to consider in interpretation of sequence analysis results, click here.

5.

Gene-targeted deletion/duplication analysis detects intragenic deletions or duplications. Methods used may include a range of techniques such as quantitative PCR, long-range PCR, multiplex ligation-dependent probe amplification (MLPA), and a gene-targeted microarray designed to detect single-exon deletions or duplications.

6.
7.

No data on detection rate of gene-targeted deletion/duplication analysis are available.

8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.

Approximately 16%-20% of individuals with a clinical diagnosis of Noonan syndrome who do not have an identified PTPN11 pathogenic variant are found to have an SOS1 pathogenic variant [Roberts et al 2007, Tartaglia et al 2007].

22.
23.

Recent reports have implicated additional genes associated with a Noonan syndrome-like phenotype in fewer than ten individuals each, including RRAS (2 probands) [Flex et al 2014] and A2ML1 (3 probands) [Vissers et al 2015].

Clinical Characteristics

Clinical Description

To date, including those with a clinical and a molecular genetic diagnosis and with an estimated incidence of 1:1000-1:2500, several thousand individuals have been identified with Noonan syndrome (NS) [Roberts et al 2013]. The following description of the phenotypic features associated with this condition is based on these reports.

Table 2.

Noonan Syndrome: Frequency of Select Features

Feature% of Persons
w/Feature
Comment
Eye anomalies95%
Short stature50%-70%For age, sex, & family background
HypotoniaMajorityCan contribute to feeding problems, speech articulation issues, & delayed attainment of gross motor milestones
Joint hyperextensibilityMajority
Pectus anomalyMajorityCharacteristic pectus deformity of the chest: pectus carinatum superiorly & pectus excavatum inferiorly
Cryptorchidism in males60%-80%
Congenital heart disease50%-80%25%-71% of affected persons have pulmonary valve stenosis, often w/pulmonary valve dysplasia.
Hearing loss40%May be sensorineural, conductive, or mixed
Hypertrophic cardiomyopathy10%-29%Half of those w/hypertrophic cardiomyopathy are diagnosed by age 6 mos.
Learning disability25%~10%-15% of those w/NS require special education.
Intellectual disability 16%-23%
Renal anomalies11%Most commonly dilatation of the renal pelvis
Abnormal bleeding or bruisingBleeding: 6%-10%; bruising: majority
1.

Defined as IQ <70

Prenatal features. Advanced paternal age has been observed in cohorts with simplex NS. Common perinatal findings [Stuurman et al 2019]:

  • Polyhydramnios
  • Lymphatic dysplasia including increased distended jugular lymphatic sacs, nuchal translucency, cystic hygroma, pleural effusion, and ascites
  • Relative macrocephaly
  • Cardiac and renal anomalies

In chromosomally normal fetuses with increased nuchal translucency, it is estimated that 3%-15% have PTPN11-associated NS [Stuurman et al 2019].

Growth. Birth weight is usually normal, although edema may cause a transient increase. Infants with NS frequently have feeding difficulties. This period of failure to thrive is self limited, although poor weight gain may persist for up to 18 months.

Length at birth is usually normal. Postnatal growth failure is often obvious from the first year of life [Seo & Yoo 2018]. Mean height then follows the third centile from ages two to four years until puberty, when below-average growth velocity and an attenuated adolescent growth spurt tend to occur. As bone maturity is usually delayed, prolonged growth into the 20s is possible.

Final adult height approaches the lower limit of normal: 161-167 cm in males and 150-155 cm in females. Growth curves have been developed from these cross-sectional retrospective data. One study suggests that 30% of affected individuals have height within the normal adult range, while more than 50% of females and nearly 40% of males have an adult height below the third centile [Seo & Yoo 2018].

In many affected persons, decreased IGF-I- and IGF-binding protein 3, together with low responses to provocation, suggest impaired growth hormone release or disturbance of the growth hormone / insulin-like growth factor I axis. Mild growth hormone resistance related to a post-receptor signaling defect (which may be partially compensated for by elevated growth hormone secretion) is reported in individuals with NS and a PTPN11 pathogenic variant [Seo & Yoo 2018].

Growth hormone (GH) therapy has been used in individuals with NS (see Management):

  • In Europe, GH treatment is the standard of care for children with abnormalities of the GH-IGF-I axis and could be used when GH physiology is normal.
  • Short- and long-term studies of GH treatment have been published [Ogawa et al 2004, Osio et al 2005, Noordam 2007, Noordam et al 2008, Romano et al 2009, Rohrer et al 2020] and demonstrate a consistent and significant increase in height velocity in children with Noonan syndrome who have been treated.
  • The increase in height standard deviation (SD) varies from 0.6 to 1.8 SD and may depend on age at start of treatment, duration of study, age at onset of puberty, and/or GH sensitivity [Seo & Yoo 2018].
  • Studies have shown that children with prepubertal NS growth hormone deficiency have increased their growth rate with GH therapy at a rate equivalent to girls with Turner syndrome but at a lower rate than that seen in idiopathic GH deficiency [Lee et al 2015, Zavras et al 2015].

Cardiovascular. Significant bias in the frequency of congenital heart disease may exist because many clinicians have in the past required the presence of cardiac anomalies for diagnosis of NS. The frequency of congenital heart disease is estimated at between 50% and 80%.

  • Pulmonary valve stenosis, often with dysplasia, is the most common anomaly in NS, found in 25%-71% of affected individuals; it may be isolated or associated with other cardiovascular defects.
  • Hypertrophic cardiomyopathy is found in 10%-29% of individuals with NS. It usually presents early in life: the median age at diagnosis is five months and more than 50% of individuals with NS and hypertrophic cardiomyopathy are diagnosed by age six months [Hickey et al 2011, Wilkinson et al 2012].
  • Other structural defects frequently observed include atrial septal defects (4%-57%), ventricular septal defects (1%-14%), atrioventricular canal defects (1%-13%), mitral valve abnormalities (2%-17%), aortic coarctation (2%-9%), patent ductus arteriosus (1%-6%), and tetralogy of Fallot (1%-4%) [Linglart & Gelb 2020].
  • An electrocardiographic abnormality is documented in approximately 90% of individuals with NS and may be present without concomitant structural defects. Extreme right axis deviation with superior counterclockwise frontal QRS loop, superior or left axis deviation, left anterior hemiblock, or an RSR' pattern in lead V1 (of no clinical consequence) are common findings [Sharland et al 1992].

Psychomotor development. Early developmental milestones may be delayed, likely in part as a result of the combination of joint hyperextensibility and hypotonia. The average age for sitting unsupported is around ten months and for walking is 21 months [Pierpont 2016]. About 50% of school-age children meet diagnostic criteria for a developmental coordination disorder [Lee et al 2005a] and impaired manual dexterity is significantly correlated with verbal and nonverbal intellectual functioning [Pierpont et al 2015].

Most school-age children perform well in a normal educational setting, but 25% have learning disabilities [Lee et al 2005a] and 10%-15% require special education [van der Burgt et al 1999]. Intellectual abilities are in general mildly lowered in children with NS. IQ scores below 70 are seen in 6%-23% across studies [Pierpont et al 2015]. Studies conflict with regard to strength in verbal vs nonverbal performance and no clear pattern has emerged [Pierpont et al 2015]. There may be a specific cognitive disability, either in verbal or praxic reasoning, requiring a special academic strategy and school placement.

Articulation deficiency is common (72%) but usually responds well to speech therapy. Language delay may be related to hearing loss, perceptual motor disabilities, or articulation deficiencies. The average age at first words is around 15 months and simple two-word phrases emerge on average from age 31 to 32 months [Pierpont et al 2015].

A study of the language phenotype of children and adults with NS showed that language impairments in general are more common in NS than in the general population and a majority of children (70%) receive speech and language therapy. When language issues are present, there is a higher risk for reading and spelling difficulties [Pierpont et al 2015]. Language is significantly correlated with nonverbal cognition, hearing ability, articulation, motor dexterity, and phonologic memory. No specific aspect of language was selectively affected in those with NS.

There is emerging evidence that impairment in attention and executive functioning is one of the most common neuropsychological challenges for children with NS [Pierpont et al 2015]. Studies that rely on screening measures rather than comprehensive diagnostic assessments suggest that children with NS are at heightened risk for autism spectrum disorders; however, further research is needed [Pierpont 2016].

Psychological health. Few details of psychological health in Noonan syndrome are reported. No particular syndrome of behavioral disability or psychopathology is observed, and self-esteem is comparable to age-related peers [Lee et al 2005a]. A study of 37 individuals with a molecular genetic diagnosis of Noonan syndrome demonstrated a higher incidence of emotional dysregulation, irritability, and anxiety symptomatology compared to the general population [Alfieri et al 2021]. Noonan [2005a] documented problems in a cohort of 51 adults: depression was found in 23%, and occasional substance abuse and bipolar disease were reported. Similar findings were not reported in a large UK cohort followed over many years [Shaw et al 2007]. In one study of adults with NS, 49% reported that they had been diagnosed and treated for depression and/or anxiety [Smpokou et al 2012].

Genitourinary. Renal abnormalities, generally mild, are present in 11% of individuals with NS. Dilatation of the renal pelvis is most common. Duplex collecting systems, minor rotational anomalies, distal ureteric stenosis, renal hypoplasia, unilateral renal agenesis, unilateral renal ectopia, and bilateral cysts with scarring are reported less commonly.

Male pubertal development and subsequent fertility may be normal, delayed, or inadequate. Deficient spermatogenesis may be related to cryptorchidism, which is noted in 60%-80% of males; however, a study of male gonadal function identified Sertoli cell dysfunction both in males with cryptorchidism and those with normal testicular descent, suggesting an intrinsic defect leading to hypergonadotropic hypogonadism [Moniez et al 2018].

Puberty may be delayed in females, with a mean age at menarche of 14.6±1.17 years. Normal fertility is the rule.

Facial features. Differences in facial appearance, albeit subtle at certain ages, are a key clinical feature:

  • In the neonate, tall forehead, widely spaced eyes with downslanted palpebral fissures, low-set, posteriorly rotated ears with a thickened helix, a deeply grooved philtrum with high, wide peaks to the vermilion border of the upper lip, and a short neck with excess nuchal skin and low posterior hairline are found.
  • In infancy, eyes are prominent, with horizontal palpebral fissures, widely spaced eyes, and full or ptotic upper eyelids. The nose has a depressed nasal bridge, wide base, and bulbous tip.
  • In childhood, facial appearance is often lacking in affect or expression, as in an individual with a myopathy.
  • By adolescence, facial shape is an inverted triangle, wide at the forehead and tapering to a pointed chin. Eyes are less prominent and features are sharper. The neck lengthens, accentuating skin webbing or prominence of the trapezius muscle.
  • In the older adult, nasolabial folds are prominent, and the skin appears transparent and wrinkled.

Skeletal features

  • Thoracic scoliosis is reported in 13%-30% of individuals diagnosed at a mean age of nine years.
  • Estimates of the frequency of the characteristic appearance of the chest (superior pectus carinatum and inferior pectus excavatum with a broad chest and increased inter-nipple distance) range from 28% to 95% [Rodríguez et al 2020].
  • Vertebral defects have also been reported.
  • Reported upper limb anomalies include cubitus valgus, radioulnar synostosis, brachydactyly, and fifth finger clinodactyly.
  • Common maxillofacial features include micrognathia, high arched palate, and dental crowding [Rodríguez et al 2020].
  • Small studies have suggested lower bone mineral density in children and osteopenia in adults [Fowlkes et al 2021].
  • Multiple giant cell lesions of the jaw, joints (pigmented villonodular synovitis), and/or soft tissue have been reported in association with PTPN11-, SOS1-, and RAF1-associated cases of Noonan syndrome [Miri et al 2018, Rodríguez et al 2020].

Bleeding diathesis. Most persons with NS have a history of abnormal bleeding or bruising. Early studies reported that about one third of all individuals with NS have one or more coagulation defects, with subsequent studies suggesting a lower rate of coagulopathy [Derbent et al 2010]. The coagulopathy may manifest as severe surgical hemorrhage, clinically mild bruising, or laboratory abnormalities with no clinical consequences. A variety of small studies have shown that while 50%-89% of those with NS have either a history of bleeding and/or abnormal hemostatic lab results, only 10%-42% have both (reviewed in Briggs & Dickerman [2012]). A study of 70 individuals with NS who had not had preoperative evaluation for coagulopathy demonstrated a 6.2% risk of perioperative bleeding complications [Briggs et al 2020].

Lymphatic. Varied lymphatic abnormalities are described in individuals with NS. They may be localized or widespread, prenatal, and/or postnatal. Dorsal limb (top of the foot and back of the hand) lymphedema is most common. Less common findings include: intestinal, pulmonary, or testicular lymphangiectasia; chylous effusions of the pleural space and/or peritoneum; and localized lymphedema of the scrotum or vulva.

Ocular. Ocular abnormalities including ptosis, strabismus, refractive errors, amblyopia, and nystagmus occur in up to 95% of affected individuals. Anterior segment and fundus changes are less common. There are case reports of keratoconus and Axenfeld anomaly [Lee & Sakhalkar 2014, Guerin et al 2015, van Trier et al 2018].

Ears/hearing. Hearing impairment has an estimated incidence of 40%. Some individuals have sensorineural hearing loss, others have a secondary conductive hearing loss due to chronic otitis media or middle ear effusion, and some have mixed conductive and sensorineural hearing loss [van Nierop et al 2017].

Dermatologic. Skin differences, particularly follicular keratosis over extensor surfaces and face, are relatively common and may occasionally be as severe as those found in cardiofaciocutaneous syndrome (see Differential Diagnosis).

Café au lait spots and lentigines are described in NS more frequently than in the general population (see Noonan syndrome with multiple lentigines discussion in Genetically Related Disorders).

Other

  • Arnold-Chiari I malformation. Eleven cases of Arnold-Chiari malformation have been reported in the medical literature; the true incidence in NS is not known [Smpokou et al 2012, Keh et al 2013, Mitsuhara et al 2014, Zarate et al 2014, Ejarque et al 2015].
  • Hepatosplenomegaly is frequent; the cause is likely related to subclinical myelodysplasia.
  • Juvenile myelomonocytic leukemia (JMML). The most common hematologic disorder in NS is transient myeloproliferative disorder, typically diagnosed in the neonatal period or early infancy. An estimated 10% of these cases progress to JMML [Niemeyer 2014]. Individuals with Noonan syndrome and a germline pathogenic variant in PTPN11 have a predisposition to this unusual childhood leukemia. In general, JMML in Noonan syndrome runs a more benign course. The associated variants are different from the somatic pathogenic variants in PTPN11-associated JMML, which ‒ when present as germline variants ‒ are associated with neonatal-lethal NS [Mason-Suares et al 2017].
  • Other malignancies. One study of individuals with Noonan syndrome caused by a pathogenic variant in PTPN11 supports a threefold increased risk of malignancy [Jongmans et al 2011].
  • Noonan-like / multiple giant-cell lesion syndrome. The giant-cell granulomas and bone and joint anomalies in Noonan-like / multiple giant-cell lesion syndrome are recognized to be part of the Noonan syndrome spectrum. They can resemble cherubism (an autosomal dominant disorder caused by pathogenic variants in SH3BP2; see Cherubism), lesions observed in neurofibromatosis (see Neurofibromatosis Type 1), or lesions observed in the Ramon syndrome with juvenile rheumatoid arthritis (polyarticular pigmented villonodular synovitis).
    Noonan-like / multiple giant-cell lesion syndrome is caused by pathogenic variants in PTPN11 [Jafarov et al 2005, Wolvius et al 2006] and SOS1 [Beneteau et al 2009, Neumann et al 2009]. One family with Noonan-like / multiple giant-cell lesion syndrome has a PTPN11 pathogenic variant that was also reported in Noonan syndrome without giant-cell lesions [Tartaglia et al 2002]; thus, additional genetic factors may be necessary for the giant-cell proliferation to occur.
  • These multiple giant cell lesions are also recognized in persons with cardiofaciocutaneous syndrome, caused by mutation of BRAF and MEK1 [Neumann et al 2009]. Thus, dysregulation of the RAS-MAPK pathway represents the common and basic molecular event predisposing to giant-cell lesion formation, arguing against the existence of Noonan-like / multiple giant-cell lesion syndrome as a separate entity.
  • Overall risk of malignancy. Kratz et al [2015] reported on a cohort of 632 individuals with molecularly confirmed NS (inclusive of Noonan syndrome with multiple lentigines) and found four individuals with JMML, two with brain tumor, two with ALL, and one with neuroblastoma; they calculated a childhood cancer standardized incidence ratio of 8.1 [Kratz et al 2015]. Individuals with NS are at an eightfold greater risk of developing a childhood cancer than are those without NS. The overall cancer risk by age 20 years is estimated to be 4% [Lodi et al 2020].
    An international meeting of the American Association for Cancer Research was held to propose consensus surveillance recommendations for RASopathies and other genetic disorders associated with increased childhood cancer risk. Overall, because the cancer risk falls below 5%, routine cancer surveillance was not recommended. However, for those with PTPN11 or KRAS variants known to be associated with myeloproliferative disorder or JMML, physical examination every three to six months with spleen size assessment and complete blood count from birth/time of diagnosis to age five years was recommended for consideration ‒ though there is not yet evidence that this strategy leads to survival advantage [Villani et al 2017].

Phenotype Correlations by Gene

There are no known phenotype correlations for RASA2 or RRAS2.

Table 3.

Noonan Syndrome Phenotype Correlations by Gene

Gene 1Phenotypic Feature
HCMEctodermal
findings
IDPulmonary
stenosis
Other
BRAF +++ 2Typically classic features
KRAS +++ 3Craniosynostosis reported in 2 persons 4
LZTR1 ++ 5
MAP2K1 +++ 2Typically classic features
MRAS +++6 of 6 reported persons 6
PTPN11 7+++Persons are also more likely to have short stature, pectus anomaly, easy bruising, cryptorchidism, & characteristic facial features.
RAF1 +++ 8++ 9
RIT1 ++ 10++ 1112Persons are less likely to have short stature & pectus anomalies & more likely to have perinatal abnormalities, high birth weight, & relative macrocephaly.
SOS1 ++-Persons are more likely to have normal stature & cardiac septal defects.
SOS2 ++-Infrequent short stature; lymphatic abnormalities in >50% 13

+, ++, +++ = degree of likelihood that a given feature is present (i.e., more +s = more likely); - = a feature that is less likely to be present. Blank cells indicate that there is no known correlation between the likelihood of having a particular feature when a person has a pathogenic variant in that gene.

HCM = hypertrophic cardiomyopathy; ID = intellectual disability

1.

Genes are listed in alphabetic order.

2.
3.

Those with pathogenic variants in KRAS tend to have a greater likelihood and severity of intellectual disability [Zenker et al 2007].

4.
5.

In one study, 5/26 persons with a heterozygous pathogenic variant in LZTR1 and19/26 with biallelic pathogenic variants in LZTR1 had hypertrophic cardiomyopathy.

6.
7.

Those with a pathogenic variant in PTPN11 are less likely to have hypertrophic cardiomyopathy compared to those with Noonan syndrome of other genetic causes.

8.

The chance of having hypertrophic cardiomyopathy in persons with Noonan syndrome caused by a heterozygous pathogenic variant in RAF1 is ~85% compared to 18% for those with Noonan syndrome in general [Gelb et al 2015].

9.

Multiple nevi, lentigines, and/or café au lait spots were reported in one third of those with RAF1-associated NS.

10.

Hypertrophic cardiomyopathy is present in 70%-75% of persons with RIT1-associated NS [Aoki et al 2013, Yaoita et al 2016].

11.

Including curly hair, hyperpigmentation, and wrinkled palms and soles

12.

Those with RIT1-NS are less likely to have intellectual disability [Bertola et al 2014, Yaoita et al 2016].

13.

Genotype-Phenotype Correlations

No genotype-phenotype correlations for BRAF, KRAS, LZTR1, MAP2K1, MRAS, NRAS, RAF1, RASA2, RIT1, RRAS2, SOS1, or SOS2 have been identified.

PTPN11

  • Germline pathogenic variants at codons 61, 71, 72, and 76 are significantly associated with leukemogenesis and identify a subgroup of individuals with NS at risk for JMML [Niihori et al 2005].
  • Individuals with the p.Asn308Asp pathogenic variant are said to be more likely to receive a typical education [Jongmans et al 2004].
  • An in-frame three-nucleotide PTPN11 deletion (p.Gly60del) in a female infant with severe features of Noonan syndrome, including hydrops fetalis and juvenile myelomonocytic leukemia [Yoshida et al 2004], has been reported. The p.Asp61del three-nucleotide PTPN11 deletion has also been reported in a child with typical rather than severe NS [Lee et al 2005b].

Nomenclature

An early term for NS, "male Turner syndrome," incorrectly implied that the condition would not be found in females.

In 1949, Otto Ullrich reported affected individuals and noted a similarity between their features and those in a strain of mice bred by Bonnevie (webbed neck and lymphedema). The term "Bonnevie-Ullrich syndrome" became popular, particularly in Europe.

Prevalence

NS is common and reported to occur in between 1:1,000 and 1:2,500 persons. Mild expression is likely to be overlooked.

Differential Diagnosis

Turner syndrome, typically seen in females, is differentiated from Noonan syndrome (NS) by demonstration of a sex chromosome abnormality on cytogenetic studies in affected individuals. The phenotype of Turner syndrome is quite different from that of NS, when one considers face, heart, development, and kidneys. In Turner syndrome, renal anomalies are more common, developmental delay is much less frequently found, and left-sided heart defects are the rule.

Genes of interest in the differential diagnosis of NS are summarized in Table 5.

Table 5.

Genes of Interest in the Differential Diagnosis of Noonan Syndrome

Gene(s)DisorderMOIClinical Characteristics / Comment
BRAF
KRAS
MAP2K1
MAP2K2 1
Cardiofaciocutaneous syndrome ADSee Genetically Related Disorders.
BRAF
MAP2K1
PTPN11
RAF1
Noonan syndrome w/multiple lentigines (previously referred to as LEOPARD syndrome)ADSee Genetically Related Disorders.
CBL 2Noonan syndrome-like disorder ± juvenile myelomonocytic leukemia (OMIM 613563)ADVariable phenotype characterized by relatively high frequency of neurologic features, predisposition to JMML, low prevalence of cardiac defects, ↓ growth, & cryptorchidism 3
FGD1 X-linked Aarskog syndrome (OMIM 305400)XLCharacterized by DD, short stature, congenital heart defects, & distinctive facies
HRAS Costello syndrome ADTypically characterized by failure to thrive in infancy due to severe postnatal feeding difficulties; short stature; DD or ID; coarse facial features; curly or sparse, fine hair; loose, soft skin w/deep palmar & plantar creases; papillomata of the face & perianal region; diffuse hypotonia & joint laxity w/ulnar deviation of wrists & fingers; tight Achilles tendons; & cardiac involvement. Relative or absolute macrocephaly is typical.
NF1 Neurofibromatosis 1 (NF1)ADNF1 shares some features w/NS, incl short stature, learning difficulties, & café au lait patches. Rare affected persons may have a NS-like facial appearance. 4
Watson syndrome, an NF1 variant, is characterized by multiple café au lait spots, pulmonic stenosis, ID, & short stature.
PPP1CB
SHOC2 2
Noonan syndrome w/loose anagen hair (OMIM PS607721)ADNS-like features or, in a small proportion of affected persons, the classic NS phenotype5 The recurrent pathogenic missense SHOC2 variant 4A>G has been found in a subgroup w/features of NS but also GH deficiency; distinctive hyperactive behavior that improves w/age in most; hair anomalies; darkly pigmented skin w/eczema or ichthyosis; hypernasal voice; & overrepresentation of mitral valve dysplasia & septal defects in comparison w/classic NS 1, 6
SPRED1 Legius syndrome ADMajority: café au lait macules, 30%-50% skin freckling, 30% developmental issues, 15% Noonan-like facial features

AD = autosomal dominant; DD = developmental delay; GH = growth hormone; GI = gastrointestinal; ID = intellectual disability; JMML = juvenile myelomonocytic leukemia; MOI = mode of inheritance; NS = Noonan syndrome; XL = X-linked

1.
2.

Due to the significant phenotypic overlap with classic NS, most RASopathy diagnostic gene panels include testing for the common SHOC2 variant and CBL gene sequencing.

3.
4.

This could be caused by chance concurrence of NS and NF1 [Colley et al 1996, Bertola et al 2005]. However, most often it appears to be a NS-like facial appearance in a person with a pathogenic variant in NF1, sometimes in the presence of a variant NF1 phenotype [Stevenson et al 2006, Nyström et al 2009].

5.

B Kerr, personal observation

6.

Other. NS should be distinguished from other syndromes/conditions with developmental delay, short stature, congenital heart defects, and distinctive facies, especially the following:

  • Autosomal dominant Aarskog syndrome of unknown genetic cause (OMIM 100050)
  • In utero exposure to alcohol or primidone

Management

Management guidelines have been developed by DYSCERNE, a European consortium [Noonan Syndrome Guideline Development Group 2010] (full text); a separate set has been published by an American consortium working with the Noonan Syndrome Support Group [Romano et al 2010] and in the Lancet [Roberts et al 2013].

Evaluations Following Initial Diagnosis

To establish the extent of disease and needs in an individual diagnosed with Noonan syndrome, the evaluations summarized in Table 6 (if not performed as part of the evaluation that led to the diagnosis) are recommended.

Table 6.

Recommended Evaluations Following Initial Diagnosis in Individuals with Noonan Syndrome

System/ConcernEvaluationComment
Constitutional Measurements of growth parameters
  • On NS-specific growth charts
  • To identify those w/failure to thrive &/or short stature
Endocrine Bone age, growth hormone & thyroid function studies 1In children w/short stature (height >2 SD below standard growth curve or crossing 2 major height %iles)
Gastrointestinal/
Feeding
Gastroenterology / nutrition / feeding team eval
  • In infants w/poor weight gain, dysphagia
  • Eval for malrotation if persistent unexplained vomiting
Development Developmental assessmentTo incl:
  • Motor, adaptive, cognitive, & speech-language eval
  • Eval for early intervention / special education
Psychiatric/
Behavioral
Neuropsychiatric eval may be considered.For those age >12 mos: screen for behavior concerns, autism, depression, ADHD, & anxiety (some symptoms may not be present until adulthood).
Cardiovascular Echocardiogram & EKGTo identify congenital heart defects, cardiomyopathy, &/or cardiac conduction abnormalities
Genitourinary Kidney ultrasoundTo assess for renal anomalies; if present, referral to urologist
Assessment for cryptorchidism in malesReferral to urologist if cryptorchidism is present
Musculoskeletal PT/OT evalTo incl:
  • Assessment of gross motor & fine motor skills
  • Need for PT (to improve gross motor skills) &/or OT (to improve fine motor skills)
Consider radiographs of the spine if asymmetry or scoliosis is present on physical exam.Consider referral to orthopedist.
Hematologic/
Lymphatic
In consultation w/hematologist: bleeding history, CBC w/differential, PT/aPTT, factor XI, XII, IX, VIII, vWf, & platelet aggregation testingIf initial screening was performed before age 12 mos, repeat after age 12 mos. 2
Eyes Ophthalmologic evalTo assess for amblyopia, refractive error, nystagmus, strabismus, & clinically significant ptosis
Hearing Audiologic evalAssess for hearing loss & middle ear effusion.
Integument Full skin examConsider referral to dermatologist in those w/multiple lentigines requiring monitoring or significant xeroderma. 3
Neurologic Neurologic evalTo incl brain & spine MRI if signs or symptoms consistent w/possible Chiari malformation
Genetic
counseling
By genetics professionals 4To inform affected persons & their families re nature, MOI, & implications of NS in order to facilitate medical & personal decision making
Family support
& resources
Assess need for:

ADHD = attention-deficit/hyperactivity disorder; MOI = mode of inheritance; NS = Noonan syndrome; OT = occupational therapy; PT = physical therapy; PT/aPTT = prothrombin/activated partial thromboplastin time; vWF = von Willebrand factor

1.

Thyroid function tests may include TSH and free T4.

2.
3.
4.

Medical geneticist, certified genetic counselor, certified advanced genetic nurse

Treatment of Manifestations

Table 7.

Treatment of Manifestations in Individuals with Noonan Syndrome

Manifestation/
Concern
TreatmentConsiderations/Other
Short stature GH therapy may be considered.
  • No standard dose has been established.
  • No apparent correlation between dosage used & final height, though earlier age at initiation of GH therapy is assoc w/↑ final adult height.
  • Short stature due to NS is an FDA-approved indication for GH treatment.
  • No evidence supports ↑ prevalence of neoplasm, cardiac, or other comorbidities in those treated w/GH. 1
Feeding
difficulties
Consideration of nasogastric tube feedings in infants w/poor growth, esp if they have a congenital heart defect or cardiomyopathyInvasive intervention (i.e., placement of gastrostomy tube) may be needed, though feeding issues are often self-limited.
DD/ID See Developmental Delay / Intellectual Disability Management Issues.
Psychiatric/
Behavioral
Standard treatment for ADHD & any neuropsychiatric features &/or behavioral problemsReferral to a neurodevelopmental specialist or psychiatrist, depending on age, may be considered.
Congenital heart defects Standard treatment per cardiologistPulmonary valve stenosis treated w/percutaneous balloon pulmonary valvuloplasty has a higher reintervention rate vs pulmonary valve stenosis w/o NS but is still considered 1st-line treatment. 2
HCM Standard treatment per cardiologistHCM is assoc w/substantial early mortality; infants presenting before age 6 mos in congestive heart failure have worst prognosis (2-yr survival of 30%). 3
Cryptorchidism Standard treatment per urologist
Renal anomalies /
Hydronephrosis
Standard treatment per urologist &/or nephrologist
Bleeding
diathesis
Standard treatment per hematologist
  • Specific treatment for serious bleeding may be guided by knowledge of a factor deficiency or platelet aggregation anomaly.
  • Factor VIIa has also been used in an infant w/NS who had normal platelet count & prothrombin & partial thromboplastin times to control severe postoperative blood loss due to gastritis. 4
Abnormal
vision &/or
strabismus
Standard treatment(s) as recommended by ophthalmologistCommunity vision services through early intervention or school district
Hearing Hearing aids may be helpful; as per otolaryngologist.Community hearing services through early intervention or school district
Chiari
malformation
Standard treatment per neurosurgeon
Juvenile
myelomonocytic
leukemia & other
malignancies
Standard treatment per oncologist
Family/
Community
  • Ensure appropriate social work involvement to connect families w/local resources, respite, & support.
  • Coordinate care to manage multiple subspecialty appointments, equipment, medications, & supplies.
  • Ongoing assessment of need for palliative care involvement &/or home nursing
  • Consider involvement in adaptive sports or Special Olympics.

ADHD = attention-deficit/hyperactivity disorder; DD/ID = developmental delay / intellectual disability; GH = growth hormone; HCM = hypertrophic cardiomyopathy; NS = Noonan syndrome

1.
2.
3.
4.

Developmental Delay / Intellectual Disability Management Issues

The following information represents typical management recommendations for individuals with developmental delay / intellectual disability in the United States; standard recommendations may vary from country to country.

Ages 0-3 years. Referral to an early intervention program is recommended for access to occupational, physical, speech, and feeding therapy as well as infant mental health services, special educators, and sensory impairment specialists. In the US, early intervention is a federally funded program available in all states that provides in-home services to target individual therapy needs.

Ages 3-5 years. In the US, developmental preschool through the local public school district is available for those who qualify. Before placement, an evaluation is made to determine needed services and therapies and an individualized education plan (IEP) is developed for those who qualify based on established motor, language, social, or cognitive delay. The early intervention program typically assists with this transition. Developmental preschool is center based.

All ages. Consultation with a developmental pediatrician may be considered to ensure the involvement of appropriate community, state, and educational agencies (US) and to support parents in maximizing quality of life. Some issues to consider:

  • IEP services:
    • An IEP provides specially designed instruction and related services to children who qualify.
    • IEP services will be reviewed annually to determine whether any changes are needed.
    • Special education law requires that children participating in an IEP be in the least restrictive environment feasible at school and included in general education as much as possible, when and where appropriate.
    • Vision and hearing consultants should be a part of the child's IEP team to support access to academic material.
    • PT, OT, and speech services will be provided in the IEP to the extent that the need affects the child's access to academic material. Beyond that, private supportive therapies based on the affected individual's needs may be considered. Specific recommendations regarding type of therapy can be made by a developmental pediatrician, as needed.
    • As a child enters the teen years, a transition plan should be discussed and incorporated in the IEP. For those receiving IEP services, the public school district is required to provide services until age 21.
  • A 504 plan (Section 504: a US federal statute that prohibits discrimination based on disability) can be considered for those who require accommodations or modifications such as front-of-class seating, assistive technology devices, classroom scribes, extra time between classes, modified assignments, and enlarged text.
  • Developmental Disabilities Administration (DDA) enrollment may be considered. DDA is a US public agency that provides services and support to qualified individuals. Eligibility differs by state but is typically determined by diagnosis and/or associated cognitive/adaptive disabilities.
  • Families with limited income and resources may also qualify for supplemental security income (SSI) for their child with a disability.

Surveillance

Table 8.

Recommended Surveillance for Individuals with Noonan Syndrome

System/ConcernEvaluationFrequency
Constitutional
  • Measurement of growth parameters on NS-specific growth charts
  • Eval of nutritional status in infants & toddlers
At each visit
Development Monitor developmental progress & educational needs.
Neurologic Assess for new manifestations, e.g., chronic headache, neck pain, changes in tone, dizziness, or signs/symptoms of OSA that may indicate a Chiari malformation. 1
Integument Skin exam
Family/
Community
Assess family need for social work support (e.g., palliative/respite care, home nursing, other local resources) & care coordination.
Psychiatric/
Behavioral
Behavioral assessment for anxiety, attention, & depressionAt each visit starting in early childhood, as age appropriate
Cardiovascular In children <5 yrs: if initial cardiac eval is normalAt least annual cardiac eval until age 5 yrs or as clinically indicated
In children >5 yrs through adulthoodAt least every 5 yrs or as clinically indicated
Eyes Ophthalmology evalAnnually in childhood & adolescence or as clinically indicated
Hearing Audiology evalAnnually in early childhood or as clinically indicated
Malignancy/
JMML 2
For those w/pathogenic PTPN11 or KRAS variants 3: consider physical exam w/assessment of spleen size & CBC.Every 3-6 mos until age 5 yrs
Coagulation/
Bleeding
In consultation w/hematologist: bleeding history, CBC w/differential, PT/aPTT, factor XI, XII, IX, VIII, vWf, & platelet aggregation testingPrior to any surgical procedure or if there is a bleeding history

CBC = complete blood count; PT/aPTT = prothrombin/activated partial thromboplastin time; vWF = von Willebrand factor; JMML = juvenile myelomonocytic leukemia; OSA = obstructive sleep apnea

1.

With referral to neurologist and consideration of head MRI to include sections through the base of the skull to assess for Chiari malformation

2.

Despite the apparent increased incidence of hematologic and solid tumor malignancies, no consensus surveillance strategies have been evaluated or recommended.

3.

Agents/Circumstances to Avoid

Aspirin therapy should be avoided because it may exacerbate a bleeding diathesis.

Evaluation of Relatives at Risk

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

Pregnancy Management

For affected women who are pregnant, consider referral to an adult congenital heart program for peripartum evaluation and management.

Pregnancy is generally a time of increased coagulation, but consider a hematology referral if the pregnant woman has a history of bleeding abnormalities and/or has not undergone previous screening for coagulopathy.

Therapies Under Investigation

The MEK inhibitor trametinib was given under compassionate use to two infants with pathogenic variants in RIT1 and progressive, congenital hypertrophic cardiomyopathy. Trametinib is a highly selective reversible allosteric inhibitor of MEK1/2 activity. In both cases, there was reversal of progressive myocardial hypertrophy and valvar obstruction along with a catch-up pattern of somatic growth [Andelfinger et al 2019].

Dori et al [2020] reported an individual age 14 years with SOS1-related NS who had resolution of mesenteric and retroperitoneal lymphangiectasia and chylothorax after treatment with trametinib, with complete remodeling of the lymphatic system.

Andrew Dauber, MD, Children's National Research Institute, is enrolling children with genetic causes of short stature, including Noonan syndrome, for a vosoritide treatment trial. Vasoritide is a selective NPR-B agonist that targets the growth plate. See Clinical Trials.

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

Noonan syndrome (NS) caused by pathogenic variants in BRAF, KRAS, MAP2K1, MRAS, NRAS, PTPN11, RAF1, RASA2, RIT1, RRAS2, SOS1, or SOS2 is inherited in an autosomal dominant manner.

NS caused by pathogenic variants in LZTR1 can be inherited in an autosomal dominant or autosomal recessive manner.

Autosomal Dominant Inheritance ‒ Risk to Family Members

Parents of a proband

  • 30%-75% of individuals diagnosed with NS have an affected parent. (Note: Because NS is associated with variable expressivity and the manifestations of the disorder are frequently subtle, many affected adults are diagnosed only after the birth of a more obviously affected infant.)
  • A proband with NS may have the disorder as the result of a de novo pathogenic variant in an NS-related gene.
  • In simplex cases (i.e., those with no known family history), paternal origin of the de novo pathogenic variant has been found universally to date [Yoon et al 2013]. In this cohort, advanced paternal age was observed along with a significant sex-ratio bias favoring transmission to males, a finding that is thus far unexplained.
  • If the proband is the only family member known to have NS, recommended evaluations of both parents include the following:
    • A thorough physical examination with particular attention to the features of NS; echo- and electrocardiography; coagulation screening; and examination of photographs of the face at all ages for characteristic features of NS
    • Molecular genetic testing if the NS-causing pathogenic variant in the proband is known
  • If the proband has an NS-causing pathogenic variant that is not identified in either parent and parental identity testing has confirmed biological maternity and paternity, the following possibilities should be considered:
  • The family history of some individuals diagnosed with NS may appear to be negative because of failure to recognize the disorder in affected family members. Therefore, an apparently negative family history cannot be confirmed without appropriate clinical evaluation of the parents and/or molecular genetic testing (to establish that neither parent is heterozygous for the pathogenic variant identified in the proband).

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

  • If a parent is affected and/or is known to have the pathogenic variant identified in the proband, the risk to the sibs is 50%. Because there can be significant intrafamilial variability, sibs may not have the same phenotypic findings as the proband.
  • If the parents are clinically unaffected and the proband has an NS-causing 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 (germline mosaicism for the PTPN11 c.922A>G pathogenic variant has been reported [Yoon et al 2013]).

Offspring of a proband. Each child of an individual with NS has a 50% chance of inheriting the NS-related pathogenic variant.

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 is known to have the familial pathogenic variant, the parent's family members may be at risk.

Autosomal Recessive Inheritance ‒ Risk to Family Members

Parents of a proband

Sibs of a proband

  • If both parents are known to be heterozygous for an LZTR1 pathogenic variant, each sib of an affected individual has at conception a 25% chance of inheriting two pathogenic variants and being affected, a 50% chance of inheriting one LZTR1 pathogenic variant and being heterozygous, and a 25% chance of inheriting neither familial pathogenic variant.
  • Heterozygotes may be asymptomatic [Johnston et al 2018] or may have mild features of NS [Jenkins et al 2020].

Offspring of a proband. The offspring of an individual with NS are obligate heterozygotes for a pathogenic variant in LZTR1.

Other family members. Each sib of the proband's parents is at a 50% risk of being heterozygous for an LZTR1 pathogenic variant.

Heterozygote Detection

Heterozygote detection for at-risk relatives requires prior identification of the LZTR1 pathogenic variants in the family.

Related Genetic Counseling Issues

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.

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

High a priori-risk pregnancy

  • Molecular genetic testing. Once the NS-related pathogenic variant(s) have been identified in an affected family member, prenatal and preimplantation genetic testing are possible.
  • Ultrasound examination. For a pregnancy at 50% risk, high-resolution ultrasound examination is also possible. Prenatal features are nonspecific but may include polyhydramnios, hydronephrosis, pleural effusion, edema, cardiac defects, distended jugular lymphatic sacs, cystic hygroma, and increased nuchal translucency [Myers et al 2014]. Cystic hygroma may be accompanied by scalp edema, polyhydramnios, pleural and pericardial effusions, ascites, and/or frank hydrops fetalis. The presence of these findings should suggest the diagnosis of NS. In addition, a search for a cardiac defect should be made; congenital heart disease is diagnosed prenatally in about 20% of cases [Myers et al 2014]. Treatment of these pregnancy complications is the same as that in the general population.

Low a priori-risk pregnancy (i.e., a fetus at no known increased risk of NS). Although the ultrasonographic findings described suggest the diagnosis of NS in high-risk pregnancies, they are nonspecific and may be associated with cardiovascular defects or other chromosome and non-chromosome syndromes. In a retrospective analysis of 309 pregnancies with isolated increased nuchal translucency (>99%ile) and no other reported ultrasound findings, four pregnancies were subsequently diagnosed with NS [Pauta et al 2022]. Of 44 pregnancies with nonimmune hydrops fetalis enrolled in the Hydrops-Yielding Diagnostic Results of Prenatal Sequencing (HYDROPS) study, four were diagnosed with NS by trio exome analysis [Al-Kouatly et al 2021].

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.

Noonan Syndrome: Genes and Databases

Data are compiled from the following standard references: gene from HGNC; chromosome locus from OMIM; protein from UniProt. For a description of databases (Locus Specific, HGMD, ClinVar) to which links are provided, click here.

Table B.

OMIM Entries for Noonan Syndrome (View All in OMIM)

163950NOONAN SYNDROME 1; NS1
164757B-RAF PROTOONCOGENE, SERINE/THREONINE KINASE; BRAF
164760RAF1 PROTOONCOGENE, SERINE/THREONINE KINASE; RAF1
164790NRAS PROTOONCOGENE, GTPase; NRAS
176872MITOGEN-ACTIVATED PROTEIN KINASE KINASE 1; MAP2K1
176876PROTEIN-TYROSINE PHOSPHATASE, NONRECEPTOR-TYPE, 11; PTPN11
182530SOS RAS/RAC GUANINE NUCLEOTIDE EXCHANGE FACTOR 1; SOS1
190070KRAS PROTOONCOGENE, GTPase; KRAS
600098RELATED RAS VIRAL ONCOGENE HOMOLOG 2; RRAS2
600574LEUCINE ZIPPER-LIKE TRANSCRIPTIONAL REGULATOR 1; LZTR1
601247SOS RAS/RAC GUANINE NUCLEOTIDE EXCHANGE FACTOR 2; SOS2
601589RAS p21 PROTEIN ACTIVATOR 2; RASA2
605275NOONAN SYNDROME 2; NS2
608435MUSCLE RAS VIRAL ONCOGENE HOMOLOG; MRAS
609591RIC-LIKE PROTEIN WITHOUT CAAX MOTIF 1; RIT1
609942NOONAN SYNDROME 3; NS3
610733NOONAN SYNDROME 4; NS4
611553NOONAN SYNDROME 5; NS5
613224NOONAN SYNDROME 6; NS6
613706NOONAN SYNDROME 7; NS7
615355NOONAN SYNDROME 8; NS8
616559NOONAN SYNDROME 9; NS9
616564NOONAN SYNDROME 10; NS10
618499NOONAN SYNDROME 11; NS11
618624NOONAN SYNDROME 12; NS12

Molecular Pathogenesis

The genes implicated in Noonan syndrome are part of or interact with the Ras/MAPK pathway. This pathway is a widely important signal transduction pathway. Growth factors, cytokines, hormones, and other extracellular ligands stimulate cell differentiation, proliferation, metabolism, and survival. Adaptor proteins are recruited and form a complex that converts inactive, GDP-bound RAS to its active GTP-bound form. This leads to downstream activation of the RAF-MEK-ERK pathway via sequential phosphorylation, culminating in activated ERK entering the nucleus and altering gene transcription. Noonan syndrome pathogenic variants usually enhance signal flow through this pathway [Roberts et al 2013].

Table 9.

Noonan Syndrome: Mechanism of Disease Causation

Gene 1MechanismComment/Reference
BRAF Gain of
function
Sarkozy et al [2009]
KRAS
LZTR1 Gain of
function
For heterozygous pathogenic variants that lead to autosomal dominant LZTR1-related Noonan syndrome [Motta et al 2019]
Loss of
function
Recessive variants typically influence protein synthesis/stability or subcellular localization [Motta et al 2019].
MAP2K1 Gain of
function
Cirstea et al [2010], Runtuwene et al [2011]
MRAS Higgins et al [2017], Motta et al [2020]
NRAS
PTPN11
RAF1 Pandit et al [2007]
RASA2 Chen et al [2014b]
RIT1 Aoki et al [2013]
RRAS Chen et al [2014b], Flex et al [2014]
SOS1 Roberts et al [2007]
SOS2 Chen et al [2014b], Yamamoto et al [2015], Lissewski et al [2021]
1.

Genes from Table 1 in alphabetic order

Table 10.

Noonan Syndrome: Notable Pathogenic Variants by Gene

Gene 1Reference SequencesDNA Nucleotide ChangePredicted
Protein Change
Comment [Reference]
PTPN11 NM_002834​.3
NP_002825​.3
c.179_181delGTGp.Gly60del Yoshida et al [2004]
c.181_183delGATp.Asp61del Lee et al [2005b]
NM_002834​.5
NP_002825​.3
c.172A>Gp.Asn58AspNS & JMML 2
c.174C>Gp.Asn58Lys
c.182A>Gp.Asp61Gly
c.184T>Gp.Tyr62Asp
c.188A>Gp.Tyr63Cys
c.214G>Ap.Ala72Thr
c.214G>Tp.Ala72Ser
c.215C>Gp.Ala72Gly
c.218C>Tp.Thr73Ile
c.227A>Tp.Glu76Val
c.228G>Tp.Glu76Asp
c.236A>Gp.Gln79Arg
c.417G>Cp.Glu139Asp
c.417G>Tp.Glu139Asp
c.794G>Ap.Arg265Gln
c.836A>Cp.Tyr279Ser
c.853T>Cp.Phe285Leu
c.922A>G 3p.Asn308Asp 3
c.923A>Gp.Asn308Ser
c.1403C>Tp.Thr468Met
c.1504T>Gp.Ser502Ala
c.1504T>Ap.Ser502Thr
c.1507G>Ap.Gly503Arg
c.1507G>Cp.Gly503Arg
c.1510A>Gp.Met504Val
RAF1 NM_002880​.4
NP_002871​.1
c.770C>Tp.Ser257Leu2 persons w/infantile fatal pulmonary hypertension 4

Variants listed in the table have been provided by the author. GeneReviews staff have not independently verified the classification of variants.

GeneReviews follows the standard naming conventions of the Human Genome Variation Society (varnomen​.hgvs.org). See Quick Reference for an explanation of nomenclature.

JMML = juvenile myelomonocytic leukemia; NS = Noonan syndrome

1.

Genes from Table 1 in alphabetic order

2.

Curated from Clinvar; included variants are those classified as pathogenic or likely pathogenic with both Noonan syndrome and JMML as listed conditions for the variant [Grant et al 2018].

3.

Less likely to be associated with learning issues [Jongmans et al 2004]

4.

Cancer and Benign Tumors

Sporadic tumors (including leukemia and solid tumors) occurring as single tumors in the absence of any other findings of Noonan syndrome may contain a somatic nucleotide variant in PTPN11, KRAS, LZTR1, MRAS, NRAS, BRAF, or MAP2K1 that is not present in the germline; thus, predisposition to these tumors is not heritable. See Catalogue of Somatic Mutations in Cancer.

  • Leukemia and solid tumors. Juvenile myelomonocytic leukemia (JMML) accounts for one third of myelodysplastic syndrome (MDS) and about 2% of leukemia. Pathogenic variants in NRAS, KRAS, and NF1 have been shown to deregulate the RAS-MAPK pathway leading to JMML in about 40% of affected individuals. Somatic pathogenic variants in exons 3 and 13 of PTPN11 have been demonstrated in 34% of individuals with JMML in one cohort [Tartaglia et al 2003, Tartaglia et al 2004, Hasle 2009].
  • Pathogenic variants in PTPN11 exon 3, were also found in 19% of children with MDS with an excess of blast cells, which often evolves into acute myeloid leukemia (AML) and is associated with poor prognosis. Nonsyndromic AML, especially the monocyte subtype FAB-MD, can be caused by PTPN11 pathogenic variants. These pathogenic variants cause a gain of function in tyrosine-protein phosphatase non-receptor type II (SHP-2), likely leading to an early initiating lesion in JMML oncogenesis with increased cell proliferation ‒ attributable in part to prolonged activation of the RAS-MAPK pathway.
  • The spectrum of leukemogenesis associated with PTPN11 pathogenic variants has been extended to include childhood acute lymphoblastic leukemia (ALL). Pathogenic variants were observed in 8% of individuals with B-cell precursor ALL, but not among children with T-lineage ALL [Tartaglia et al 2004]. Additionally, Bentires-Alj et al [2004] described SHP-2-activating PTPN11 pathogenic variants in solid tumors including breast, lung, and gastric neoplasms and neuroblastoma.
  • Somatic RAF1 nucleotide variants have only rarely been found in cancer; most of these cancer-causing variants do not cluster in the regions that are germline mutational hot spots in Noonan syndrome [Pandit et al 2007].
  • NRAS variants are commonly observed in somatic cancer; they occur in regions different from germline NRAS variants that cause Noonan syndrome.
  • MAP2K1 somatic variants have been reported in ovarian cancer [Estep et al 2007] and lung cancer [Marks et al 2008].
  • Somatic SOS1 pathogenic variants have not been found in cancer.
  • Recent studies have identified somatic RIT1 pathogenic variants in lung adenocarcinoma and myeloproliferative or mixed myelodysplastic/myeloproliferative neoplasms [Berger et al 2014, Cancer Genome Atlas Research Network 2014].
  • The somatic p.Gly248Arg pathogenic variant in LZTR1 has been identified in melanoma, glioblastoma, and colorectal cancers (COSMIC database).

Chapter Notes

Author Notes

Amy E Roberts, MD, FACMG

Department of Cardiology and Division of Genetics, Department of Pediatrics, Boston Children’s Hospital, Boston, MA

Email: amy.roberts@cardio.chboston.org

Website: www.childrenshospital.org/directory/amy-roberts

Dr Roberts is trained in both clinical genetics and pediatrics. Her research focuses on genotype phenotype correlations and gene discovery in Noonan syndrome and other RASopathies. She is a member of the Scientific Advisory Board of RASopathies Network and the Medical Advisory Board of CFC international.

Acknowledgments

Our understanding of Noonan syndrome is deeply informed by the life's work of the late Dr Jacqueline A Noonan [Gelb et al 2020], who first described the diagnosis in 1963. The author would like to acknowledge the original author of this GeneReview, Judith Allanson, and the mentorship, collaboration, and collegiality of leaders in the field, including Bruce Gelb, Bryan Hall, Maria Kontaridis, Raju Kucherlapati, Mary Ella Pierpont, Rene Pierpont, Alicia Romano, Marco Tartaglia, Ineke van der Burgt, and Martin Zenker. Finally, Dr Roberts would like to thank the leadership of the RASopathies Network and Noonan Syndrome Foundation as well as the many families with Noonan syndrome who have shared their children's experiences in clinic, in research, and at family meetings for their selflessness and advocacy without which much of this work would not have been possible.

Author History

Judith E Allanson, MD; Children's Hospital of Eastern Ontario (2001-2021)

Amy E Roberts, MD (2001-present)

Revision History

  • 16 December 2021 (ma) Comprehensive update posted live
  • 8 August 2019 (ma) Revision: pathogenic variants in LZTR1 associated with autosomal dominant and autosomal recessive Noonan syndrome
  • 25 February 2016 (ha) Comprehensive update posted live
  • 4 August 2011 (me) Comprehensive update posted live
  • 7 October 2008 (me) Comprehensive update posted live
  • 6 September 2007 (cd) Revision: mutations in RAF1 associated with Noonan syndrome
  • 22 December 2006 (cd) Revision: SOS1 mutations responsible for some cases of Noonan syndrome; clinical testing available
  • 22 May 2006 (cd) Revision: prenatal testing for Noonan syndrome caused by KRAS mutations clinically available
  • 16 May 2006 (cd) Revision: KRAS testing clinically available
  • 1 May 2006 (ja) Revision: mutations in KRAS cause Noonan syndrome
  • 9 March 2006 (me) Comprehensive update posted live
  • 17 December 2003 (me) Comprehensive update posted live
  • 15 November 2001 (me) Review posted live
  • 2 August 2001 (ja) Original submission

References

Published Guidelines / Consensus Statements

  • Noonan Syndrome Guideline Development Group. Management of Noonan syndrome – a clinical guideline (pdf). University of Manchester: DYSCERNE. Available online. 2010. Accessed 6-7-23.
  • Roberts AE, Allanson JE, Tartaglia M, Gelb BD. Noonan syndrome. Lancet. 2013;381:333-42. [PubMed]
  • Romano AA, Allanson JE, Dahlgren J, Gelb BD, Hall B, Pierpont ME, Roberts AE, Robinson W, Takemoto CM, Noonan JA. Noonan syndrome: clinical features, diagnosis, and management guidelines. Pediatrics. 2010;126:746-59. [PubMed]

Literature Cited

  • Alfieri P, Cumbo F, Serra G, Trasolini M, Frattini C, Scibelli F, Licchelli S, Cirillo F, Caciolo C, Casini MP, D'Amico A, Tartaglia M, Digilio MC, Capolino R, Vicari S. Manic and depressive symptoms in children diagnosed with Noonan syndrome. Brain Sci. 2021;11:233. [PMC free article: PMC7918671] [PubMed: 33668418]
  • Al-Kouatly HB, Makhamreh MM, Rice SM, Smith K, Harman C, Quinn A, Valcarcel BN, Firman B, Liu R, Hegde M, Critchlow E, Berger SI. High diagnosis rate for nonimmune hydrops fetalis with prenatal clinical exome from the Hydrops-Yielding Diagnostic Results of Prenatal Sequencing (HYDROPS) Study. Genet Med. 2021;23:1325-33. [PubMed: 33686258]
  • Altmüller F, Lissewski C, Bertola D, Flex E, Stark Z, Spranger S, Baynam G, Buscarilli M, Dyack S, Gillis J, Yntema HG, Pantaleoni F, van Loon RL, MacKay S, Mina K, Schanze I, Tan TY, Walsh M, White SM, Niewisch MR, García-Miñaúr S, Plaza D, Ahmadian MR, Cavé H, Tartaglia M, Zenker M. Genotype and phenotype spectrum of NRAS germline variants. Eur J Hum Genet. 2017;25:823-31. [PMC free article: PMC5520077] [PubMed: 28594414]
  • Andelfinger G, Marquis C, Raboisson MJ, Théoret Y, Waldmüller S, Wiegand G, Gelb BD, Zenker M, Delrue MA, Hofbeck M. Hypertrophic cardiomyopathy in Noonan syndrome treated by MEK-inhibition. J Am Coll Cardiol. 2019;73:2237-9. [PMC free article: PMC6916648] [PubMed: 31047013]
  • Aoki Y, Niihori T, Banjo T, Okamoto N, Mizuno S, Kurosawa K, Ogata T, Takada F, Yano M, Ando T, Hoshika T, Barnett C, Ohashi H, Kawame H, Hasegawa T, Okutani T, Nagashima T, Hasegawa S, Funayama R, Nagashima T, Nakayama K, Inoue S, Watanabe Y, Ogura T, Matsubara Y. Gain-of-function mutations in RIT1 cause Noonan syndrome, a RAS/MAPK pathway syndrome.Am J Hum Genet. 2013;93:173-80. [PMC free article: PMC3710767] [PubMed: 23791108]
  • Aoki Y, Niihori T, Inoue SI, Matsubara Y. Recent advances in RASopathies. J Hum Genet. 2016;61:33-9. [PubMed: 26446362]
  • Armour CM, Allanson JE (2008) Further delineation of cardio-facio-cutaneous syndrome: clinical features of 38 individuals with proven mutations. J Med Genet 45:249-54 [PubMed: 18039946]
  • Beneteau C, Cavé H, Moncla A, Dorison N, Munnich A, Verloes A, Leheup B (2009) SOS1 and PTPN11 mutations in five cases of Noonan syndrome with multiple giant cell lesions. Eur J Hum Genet. 17:1216-21. [PMC free article: PMC2986637] [PubMed: 19352411]
  • Bentires-Alj M, Paez JG, David FS, Keilhack H, Halmos B, Naoki K, Maris JM, Richardson A, Bardelli A, Sugarbaker DJ, Richards WG, Du J, Girard L, Minna JD, Loh ML, Fisher DE, Velculescu VE, Vogelstein B, Meyerson M, Sellers WR, Neel BG (2004) Activating mutations of the Noonan syndrome-associated SHP2/PTPN11 gene in human solid tumors and adult acute myelogenous leukemia. Cancer Res 64:8816-20 [PubMed: 15604238]
  • Berger AH, Imielinski M, Duke F, Wala J, Kaplan N, Shi GX, Andres DA, Meyerson M. Oncogenic RIT1 mutations in lung adenocarcinoma. Oncogene. 2014;33:4418-23. [PMC free article: PMC4150988] [PubMed: 24469055]
  • Bertola DR, Pereira AC, Passetti F, de Oliveira PS, Messiaen L, Gelb BD, Kim CA, Krieger JE (2005) Neurofibromatosis-Noonan syndrome: molecular evidence of the concurrence of both disorders in a patient. Am J Med Genet A. 136:242-5. [PubMed: 15948193]
  • Bertola DR, Yamamoto GL, Almeida TF, Buscarilli M, Jorge AA, Malaquias AC, Kim CA, Takahashi VN, Passos-Bueno MR, Pereira AC. Further evidence of the importance of RIT1 in Noonan syndrome. Am J Med Genet A. 2014;164A:2952-7. [PubMed: 25124994]
  • Brasil AS, Pereira AC, Wanderley LT, Kim CA, Malaquias AC, Jorge AA, Krieger JE, Bertola DR. PTPN11 and KRAS gene analysis in patients with Noonan and Noonan-like syndromes. Genet Test Mol Biomarkers. 2010;14:425-32. [PubMed: 20578946]
  • Briggs BJ, Dickerman JD. Bleeding disorders in Noonan syndrome. Pediatr Blood Cancer. 2012;58:167-72. [PubMed: 22012616]
  • Briggs B, Savla D, Ramchandar N, Dimmock D, Le D, Thornburg CD. The evaluation of hematologic screening and perioperative management in patients with Noonan syndrome: a retrospective chart review. J Pediatr. 2020;220:154-8.e6. [PubMed: 32111381]
  • Cancer Genome Atlas Research Network. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511:543-50. [PMC free article: PMC4231481] [PubMed: 25079552]
  • Capri Y, Flex E, Krumbach OHF, Carpentieri G, Cecchetti S, Lißewski C, Rezaei Adariani S, Schanze D, Brinkmann J, Piard J, Pantaleoni F, Lepri FR, Goh ES, Chong K, Stieglitz E, Meyer J, Kuechler A, Bramswig NC, Sacharow S, Strullu M, Vial Y, Vignal C, Kensah G, Cuturilo G, Kazemein Jasemi NS, Dvorsky R, Monaghan KG, Vincent LM, Cavé H, Verloes A, Ahmadian MR, Tartaglia M, Zenker M. Activating mutations in RRAS2 are a rare cause of Noonan syndrome. Am J Hum Genet. 2019;104:1223-32, [PMC free article: PMC6562003] [PubMed: 31130282]
  • Chen JL, Zhu X, Zhao TL, Wang J, Yang YF, Tan ZP. Rare copy number variations containing genes involved in RASopathies: deletion of SHOC2 and duplication of PTPN11. Mol Cytogenet. 2014a;7:28. [PMC free article: PMC4031927] [PubMed: 24739123]
  • Chen PC, Yin J, Yu HW, Yuan T, Fernandez M, Yung CK, Trinh QM, Peltekova VD, Reid JG, Tworog-Dube E, Morgan MB, Muzny DM6, Stein L, McPherson JD, Roberts AE, Gibbs RA, Neel BG, Kucherlapati R. Next-generation sequencing identifies rare variants associated with Noonan syndrome. Proc Natl Acad Sci U S A. 2014b;111:11473-8. [PMC free article: PMC4128129] [PubMed: 25049390]
  • Cirstea IC, Kutsche K, Dvorsky R, Gremer L, Carta C, Horn D, Roberts AE, Lepri F, Merbitz-Zahradnik T, König R, Kratz CP, Pantaleoni F, Dentici ML, Joshi VA, Kucherlapati RS, Mazzanti L, Mundlos S, Patton MA, Silengo MC, Rossi C, Zampino G, Digilio C, Stuppia L, Seemanova E, Pennacchio LA, Gelb BD, Dallapiccola B, Wittinghofer A, Ahmadian MR, Tartaglia M, Zenker M (2010) A restricted spectrum of NRAS mutations causes Noonan syndrome. Nat Genet 42:27-9. [PMC free article: PMC3118669] [PubMed: 19966803]
  • Colley A, Donnai D, Evans DG (1996) Neurofibromatosis/Noonan phenotype: a variable feature of type 1 neurofibromatosis. Clin Genet. 49:59-64. [PubMed: 8740913]
  • Cordeddu V, Di Schiavi E, Pennacchio LA, Ma'ayan A, Sarkozy A, Fodale V, Cecchetti S, Cardinale A, Martin J, Schackwitz W, Lipzen A, Zampino G, Mazzanti L, Digilio MC, Martinelli S, Flex E, Lepri F, Bartholdi D, Kutsche K, Ferrero GB, Anichini C, Selicorni A, Rossi C, Tenconi R, Zenker M, Merlo D, Dallapiccola B, Iyengar R, Bazzicalupo P, Gelb BD, Tartaglia M (2009) Mutation of SHOC2 promotes aberrant protein N-myristoylation and causes Noonan-like syndrome with loose anagen hair. Nat Genet. 41:1022-6. [PMC free article: PMC2765465] [PubMed: 19684605]
  • Cordeddu V, Yin JC, Gunnarsson C, Virtanen C, Drunat S, Lepri F, De Luca A, Rossi C, Ciolfi A, Pugh TJ, Bruselles A, Priest JR, Pennacchio LA, Lu Z, Danesh A, Quevedo R, Hamid A, Martinelli S, Pantaleoni F, Gnazzo M, Daniele P, Lissewski C, Bocchinfuso G, Stella L, Odent S, Philip N, Faivre L, Vlckova M, Seemanova E, Digilio C, Zenker M, Zampino G, Verloes A, Dallapiccola B, Roberts AE, Cavé H, Gelb BD, Neel BG, Tartaglia M. Activating mutations affecting the Dbl homology domain of SOS2 cause Noonan syndrome. Hum Mutat. 2015;36:1080-7. [PMC free article: PMC4604019] [PubMed: 26173643]
  • Denayer E, Devriendt K, de Ravel T, Van Buggenhout G, Smeets E, Francois I, Sznajer Y, Craen M, Leventopoulos G, Mutesa L, Vandecasseye W, Massa G, Kayserili H, Sciot R, Fryns JP, Legius E (2010) Tumor spectrum in children with Noonan syndrome and SOS1 or RAF1 mutations. Genes Chromosomes Cancer. 49:242-52. [PubMed: 19953625]
  • Derbent M, Oncel Y, Tokel K, Varan B, Haberal A, Yazici AC, Legius E, Ozbek N (2010). Clinical and hematological findings in Noonan syndrome patients with PTPN11 mutations. Am J Med Genet 152A:2768-74. [PubMed: 20954246]
  • Dori Y, Smith C, Pinto E, Snyder K, March ME, Hakonarson H, Belasco J. Severe lymphatic disorder resolved with MEK inhibition in a patient with Noonan syndrome and SOS1 mutation. Pediatrics. 2020;146:e20200167. [PubMed: 33219052]
  • Ejarque I, Millán-Salvador JM, Oltra S, Pesudo-Martínez JV, Beneyto M, Pérez-Aytés A. Arnold-Chiari malformation in Noonan syndrome and other syndromes of the RAS/MAPK pathway. Rev Neurol. 2015;60:408-12. [PubMed: 25912702]
  • Estep AL, Palmer C, McCormick F, Rauen KA (2007) Mutation analysis of BRAF, MEK1 and MEK2 in 15 ovarian cancer cell lines: implications for therapy. PLoS One. 2:e1279. [PMC free article: PMC2093994] [PubMed: 18060073]
  • Flex E, Jaiswal M, Pantaleoni F, Martinelli S, Strullu M, Fansa EK, Caye A, De Luca A, Lepri F, Dvorsky R, Pannone L, Paolacci S, Zhang SC, Fodale V, Bocchinfuso G, Rossi C, Burkitt-Wright EM, Farrotti A, Stellacci E, Cecchetti S, Ferese R, Bottero L, Castro S, Fenneteau O, Brethon B, Sanchez M, Roberts AE, Yntema HG, Van Der Burgt I, Cianci P, Bondeson ML, Cristina Digilio M, Zampino G, Kerr B, Aoki Y, Loh ML, Palleschi A, Di Schiavi E, Carè A, Selicorni A, Dallapiccola B, Cirstea IC, Stella L, Zenker M, Gelb BD, Cavé H, Ahmadian MR, Tartaglia M. Activating mutations in RRAS underlie a phenotype within the RASopathy spectrum and contribute to leukaemogenesis. Hum Mol Genet. 2014;23:4315-27. [PMC free article: PMC4103678] [PubMed: 24705357]
  • Fowlkes JL, Thrailkill KM, Bunn RC. RASopathies: The musculoskeletal consequences and their etiology and pathogenesis. Bone. 2021;152:116060. [PMC free article: PMC8316423] [PubMed: 34144233]
  • Garren B, Stephan M, Hogue JS. NRAS associated RASopathy and embryonal rhabdomyosarcoma. Am J Med Genet A. 2020;182:195-200. [PubMed: 31697451]
  • Gelb BD, Newburger JW, Roberts AE, Williams RG. In memoriam: Jaqueline A Noonan. J Am Coll Cardiol. 2020:76:1498-500.
  • Gelb BD, Roberts AE, Tartaglia M. Cardiomyopathies in Noonan syndrome and the other RASopathies. Prog Pediatr Cardiol. 2015;39:13-19. [PMC free article: PMC4568836] [PubMed: 26380542]
  • Graham JM Jr, Kramer N, Bejjani BA, Thiel CT, Carta C, Neri G, Tartaglia M, Zenker M (2009) Genomic duplication of PTPN11 is an uncommon cause of Noonan syndrome. Am J Med Genet 149A:2122-8. [PMC free article: PMC2768228] [PubMed: 19760651]
  • Grant AR, Cushman BJ, Cavé H, Dillon MW, Gelb BD, Gripp KW, Lee JA, Mason-Suares H, Rauen KA, Tartaglia M, Vincent LM, Zenker M. Assessing the gene-disease association of 19 genes with the RASopathies using the ClinGen gene curation framework. Hum Mutat. 2018;39:1485-93. [PMC free article: PMC6326381] [PubMed: 30311384]
  • Gripp KW, Aldinger KA, Bennett JT, Baker L, Tusi J, Powell-Hamilton N, Stabley D, Sol-Church K, Timms AE, Dobyns WB. A novel rasopathy caused by recurrent de novo missense mutations in PPP1CB closely resembles Noonan syndrome with loose anagen hair. Am J Med Genet A. 2016;170:2237-47. [PMC free article: PMC5134331] [PubMed: 27264673]
  • Guerin A, So J, Mireskandari K, Jougeh-Doust S, Chisholm C, Klatt R, Richer J. Expanding the clinical spectrum of ocular anomalies in Noonan syndrome: Axenfeld-anomaly in a child with PTPN11 mutation. Am J Med Genet A. 2015;167A:403-6. [PubMed: 25425531]
  • Hasle H (2009) Malignant diseases in Noonan syndrome and related disorders. Horm Res 72:8-14. [PubMed: 20029231]
  • Hastings R, Newbury-Ecob R, Ng A, Taylor R. A further patient with Noonan syndrome due to a SOS1 mutation and rhabdomyosarcoma. Genes Chromosomes Cancer. 2010;49:967-8. [PubMed: 20607846]
  • Hickey EJ, Mehta R, Elmi M, Asoh K, McCrindle BW, Williams WG, Manlhiot C, Benson L. Survival implications: hypertrophic cardiomyopathy in Noonan syndrome. Congenit Heart Dis 2011;6:41-7 [PubMed: 21269411]
  • Higgins EM, Bos JM, Mason-Suares H, Tester DJ, Ackerman JP, MacRae CA, Sol-Church K, Gripp KW, Urrutia R, Ackerman MJ. Elucidation of MRAS-mediated Noonan syndrome with cardiac hypertrophy. JCI Insight. 2017;2:e91225. [PMC free article: PMC5333962] [PubMed: 28289718]
  • Hopper RK, Feinstein JA, Manning MA, Benitz W, Hudgins L. Neonatal pulmonary arterial hypertension and Noonan syndrome: two fatal cases with a specific RAF1 mutation. Am J Med Genet A. 2015;167A:882-5. [PubMed: 25706034]
  • 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]
  • Huckstadt V, Chinton J, Gomez A, Obregon MG, Gravina LP. Noonan syndrome with loose anagen hair with variants in the PPP1CB gene: First familial case reported. Am J Med Genet A. 2021;185:1256-60. [PubMed: 33491856]
  • Jafarov T, Ferimazova N, Reichenberger E (2005) Noonan-like syndrome mutations in PTPN11 in patients diagnosed with cherubism. Clin Genet 68:190-1 [PubMed: 15996221]
  • Jenkins J, Barnes A, Birnbaum B, Papagiannis J, Thiffault I, Saunders CJ. LZTR1-related hypertrophic cardiomyopathy without typical Noonan syndrome features. Circ Genom Precis Med. 2020;13:e002690. [PubMed: 32004086]
  • Johnston JJ, van der Smagt JJ, Rosenfeld JA, Pagnamenta AT, Alswaid A, Baker EH, Blair E, Borck G, Brinkmann J, Craigen W, Dung VC, Emrick L, Everman DB, van Gassen KL, Gulsuner S, Harr MH, Jain M, Kuechler A, Leppig KA, McDonald-McGinn DM, Can NTB, Peleg A, Roeder ER, Rogers RC, Sagi-Dain L, Sapp JC, Schäffer AA, Schanze D, Stewart H, Taylor JC, Verbeek NE, Walkiewicz MA, Zackai EH, Zweier C, Zenker M, Lee B, Biesecker LG, et al. Autosomal recessive Noonan syndrome associated with biallelic LZTR1 variants. Genet Med. 2018;20:1175-85 [PMC free article: PMC6105555] [PubMed: 29469822]
  • Jongmans M, Otten B, Noordam K, van der Burgt I (2004) Genetics and variation in phenotype in Noonan syndrome. Horm Res 62 Suppl 3:56-9 [PubMed: 15539800]
  • Jongmans MC, Hoogerbrugge PM, Hilkens L, Flucke U, van der Burgt I, Noordam K, Ruiterkamp-Versteeg M, Yntema HG, Nillesen WM, Ligtenberg MJ, van Kessel AG, Kuiper RP, Hoogerbrugge N (2010) Noonan syndrome, the SOS1 gene and embryonal rhabdomyosarcoma. Genes Chromosomes Cancer. 49:635-41. [PubMed: 20461756]
  • Jongmans MC, van der Burgt I, Hoogerbrugge PM, Noordam K, Yntema HG, Nillesen WM, Kuiper RP, Ligtenberg MJ, van Kessel AG, van Krieken JH, Kiemeney LA, Hoogerbrugge N (2011) Cancer risk in patients with Noonan syndrome carrying a PTPN11 mutation. Eur J Hum Genet. 19:870-4. [PMC free article: PMC3172922] [PubMed: 21407260]
  • Jónsson H, Sulem P, Kehr B, Kristmundsdottir S, Zink F, Hjartarson E, Hardarson MT, Hjorleifsson KE, Eggertsson HP, Gudjonsson SA, Ward LD, Arnadottir GA, Helgason EA, Helgason H, Gylfason A, Jonasdottir A, Jonasdottir A, Rafnar T, Frigge M, Stacey SN, Th Magnusson O, Thorsteinsdottir U, Masson G, Kong A, Halldorsson BV, Helgason A, Gudbjartsson DF, Stefansson K. Parental influence on human germline de novo mutations in 1,548 trios from Iceland. Nature. 2017;549:519-22. [PubMed: 28959963]
  • Jung A, Bechthold S, Pfluger T, Renner C, Ehrt O. Orbital rhabdomyosarcoma in Noonan syndrome. J Pediatr Hematol Oncol. 2003;25:330-2. [PubMed: 12679651]
  • Keh YS, Abernethy L, Pettorini B. Association between Noonan syndrome and Chiari I malformation: a case-based update. Childs Nerv Syst. 2013;29:749-52. [PubMed: 23239255]
  • Khan S, McDowell H, Upadhyaya M, Fryer A. Vaginal rhabdomyosarcoma in a patient with Noonan syndrome. J Med Genet. 1995;32:743-5. [PMC free article: PMC1051679] [PubMed: 8544198]
  • Kratz CP, Franke L, Peters H, Kohlschmidt N, Kazmierczak B, Finckh U, Bier A, Eichhorn B, Blank C, Kraus C, Kohlhase J, Pauli S, Wildhardt G, Kutsche K, Auber B, Christmann A, Bachmann N, Mitter D, Cremer FW, Mayer K, Daumer-Haas C, Nevinny-Stickel-Hinzpeter C, Oeffner F, Schlüter G, Gencik M, Überlacker B, Lissewski C, Schanze I, Greene MH, Spix C, Zenker M. Cancer spectrum and frequency among children with Noonan, Costello, and cardio-facio-cutaneous syndromes. Br J Cancer. 2015;112:1392-7. [PMC free article: PMC4402457] [PubMed: 25742478]
  • Kratz CP, Zampino G, Kriek M, Kant SG, Leoni C, Pantaleoni F, Oudesluys-Murphy AM, Di Rocco C, Kloska SP, Tartaglia M, Zenker M. Craniosynostosis in patients with Noonan syndrome caused by germline KRAS mutations. Am J Med Genet A. 2009;149A:1036-40 [PubMed: 19396835]
  • Lee A, Sakhalkar MV. Ocular manifestations of Noonan syndrome in twin siblings: a case report of keratoconus with acute corneal hydrops. Indian J Ophthalmol. 2014;62:1171-3. [PMC free article: PMC4313503] [PubMed: 25579364]
  • Lee DA, Portnoy S, Hill P, Gillberg C, Patton MA (2005a) Psychological profile of children with Noonan syndrome. Dev Med Child Neurol 47:35-8 [PubMed: 15686287]
  • Lee PA, Ross JL, Pedersen BT, Kotnik P, Germak JA, Christesen HT. Noonan syndrome and Turner syndrome patients respond similarly to 4 years' growth-hormone therapy: longitudinal analysis of growth-hormone-naïve patients enrolled in the NordiNet® International Outcome Study and the ANSWER Program. Int J Pediatr Endocrinol. 2015;2015:17. [PMC free article: PMC4562101] [PubMed: 26351466]
  • Lee WH, Raas-Rotschild A, Miteva MA, Bolasco G, Rein A, Gillis D, Vidaud D, Vidaud M, Villoutreix BO, Parfait B. Noonan syndrome type I with PTPN11 3 bp deletion: structure-function implications. Proteins. 2005b;58:7-13 [PubMed: 15521065]
  • Linglart L, Gelb BD. Congenital heart defects in Noonan syndrome: diagnosis, management, and treatment. Am J Med Genet C Semin Med Genet. 2020;184:73-80. [PMC free article: PMC7682536] [PubMed: 32022400]
  • Lissewski C, Kant SG, Stark Z, Schanze I, Zenker M. Copy number variants including RAS pathway genes-how much RASopathy is in the phenotype? Am J Med Genet A. 2015;167A:2685-90. [PubMed: 25974318]
  • Lissewski C, Chune V, Pantaleoni F, De Luca A, Capri Y, Brinkmann J, Lepri F, Daniele P, Leenders E, Mazzanti L, Scarano E, Radio FC, Kutsche K, Kuechler A, Gérard M, Ranguin K, Legendre M, Vial Y, van der Burgt I, Rinne T, Andreucci E, Mastromoro G, Digilio MC, Cave H, Tartaglia M, Zenker M. Variants of SOS2 are a rare cause of Noonan syndrome with particular predisposition for lymphatic complications. Eur J Hum Genet. 2021;29:51-60. [PMC free article: PMC7852574] [PubMed: 32788663]
  • Lodi M, Boccuto L, Carai A, Cacchione A, Miele E, Colafati GS, Diomedi Camassei F, De Palma L, De Benedictis A, Ferretti E, Catanzaro G, Pò A, De Luca A, Rinelli M, Lepri FR, Agolini E, Tartaglia M, Locatelli F, Mastronuzzi A. Low-grade gliomas in patients with Noonan syndrome: case-based review of the literature. Diagnostics (Basel). 2020;10:582. [PMC free article: PMC7460327] [PubMed: 32806529]
  • Luo C, Yang YF, Yin BL, Chen JL, Huang C, Zhang WZ, Wang J, Zhang H, Yang JF, Tan ZP. Microduplication of 3p25.2 encompassing RAF1 associated with congenital heart disease suggestive of Noonan syndrome. Am J Med Genet A. 2012;158A:1918-23. [PubMed: 22786616]
  • Marks JL, Gong Y, Chitale D, Golas B, McLellan MD, Kasai Y, Ding L, Mardis ER, Wilson RK, Solit D, Levine R, Michel K, Thomas RK, Rusch VW, Ladanyi M, Pao W (2008) Novel MEK1 mutation identified by mutational analysis of epidermal growth factor receptor signaling pathway genes in lung adenocarcinoma. Cancer Res. 68:5524-8. [PMC free article: PMC2586155] [PubMed: 18632602]
  • Martinelli S, De Luca A, Stellacci E, Rossi C, Checquolo S, Lepri F, Caputo V, Silvano M, Buscherini F, Consoli F, Ferrara G, Digilio MC, Cavaliere ML, van Hagen JM, Zampino G, van der Burgt I, Ferrero GB, Mazzanti L, Screpanti I, Yntema HG, Nillesen WM, Savarirayan R, Zenker M, Dallapiccola B, Gelb BD, Tartaglia M. Heterozygous germline mutations in the CBL tumor-suppressor gene cause a Noonan syndrome-like phenotype. Am J Hum Genet. 2010;87:250-7. [PMC free article: PMC2917705] [PubMed: 20619386]
  • Martinelli S, Stellacci E, Pannone L, D'Agostino D, Consoli F, Lissewski C, Silvano M, Cencelli G, Lepri F, Maitz S, Pauli S, Rauch A, Zampino G, Selicorni A, Melançon S, Digilio MC, Gelb BD, De Luca A, Dallapiccola B, Zenker M, Tartaglia M. Molecular diversity and associated phenotypic spectrum of germline CBL mutations. Hum Mutat. 2015;36:787-96. [PubMed: 25952305]
  • Mason-Suares H, Toledo D, Gekas J, Lafferty KA, Meeks N, Pacheco MC, Sharpe D, Mullen TE, Lebo MS. Juvenile myelomonocytic leukemia-associated variants are associated with neo-natal lethal Noonan syndrome. Eur J Hum Genet. 2017;25:509-11. [PMC free article: PMC5386422] [PubMed: 28098151]
  • McFarlane J, Knight T, Sinha A, Cole T, Kiely N, Freeman R. Exostoses, enchondromatosis and metachondromatosis; diagnosis and management. Acta Orthop Belg. 2016;82:102-5. [PubMed: 26984661]
  • Miri O, Bonnet N, Lysy P, Loucheur N, Gayito R, Docquier PL. Multifocal pigmented villonodular synovitis in the Noonan syndrome. Case Rep Orthop. 2018;2018:7698052. [PMC free article: PMC6305014] [PubMed: 30631623]
  • Mitsuhara T, Yamaguchi S, Takeda M, Kurisu K. Gowers' intrasyringeal hemorrhage associated with Chiari type I malformation in Noonan syndrome. Surg Neurol Int. 2014;5:6. [PMC free article: PMC3927095] [PubMed: 24575321]
  • Moniez S, Pienkowski C, Lepage B, Hamdi S, Daudin M, Oliver I, Jouret B, Cartault A, Diene G, Verloes A, Cavé H, Salles JP, Tauber M, Yart A, Edouard T. Noonan syndrome males display Sertoli cell-specific primary testicular insufficiency. Eur J Endocrinol. 2018;179:409-18. [PubMed: 30325180]
  • Moschovi M, Touliatou V, Papadopoulou A, Mayakou MA, Nikolaidou-Karpathiou P, Kitsiou-Tzeli S. Rhabdomyosarcoma in a patient with Noonan syndrome phenotype and review of the literature. J Pediatr Hematol Oncol. 2007;29:341-4 [PubMed: 17483716]
  • Motta M, Fidan M, Bellacchio E, Pantaleoni F, Schneider-Heieck K, Coppola S, Borck G, Salviati L, Zenker M, Cirstea IC, Tartaglia M. Dominant Noonan syndrome-causing LZTR1 mutations specifically affect the Kelch domain substrate-recognition surface and enhance RAS-MAPK signaling. Hum Mol Genet. 2019;28:1007-22 [PubMed: 30481304]
  • Motta M, Sagi-Dain L, Krumbach OHF, Hahn A, Peleg A, German A, Lissewski C, Coppola S, Pantaleoni F, Kocherscheid L, Altmüller F, Schanze D, Logeswaran T, Chahrokh-Zadeh S, Munzig A, Nakhaei-Rad S, Cavé H, Ahmadian MR, Tartaglia M, Zenker M. Activating MRAS mutations cause Noonan syndrome associated with hypertrophic cardiomyopathy. Hum Mol Genet. 2020;29:1772-83. [PubMed: 31108500]
  • Myers A, Bernstein JA, Brennan ML, Curry C, Esplin ED, Fisher J, Homeyer M, Manning MA, Muller EA, Niemi AK, Seaver LH, Hintz SR, Hudgins L. Perinatal features of the RASopathies: Noonan syndrome, cardiofaciocutaneous syndrome and Costello syndrome. Am J Med Genet A. 2014;164A:2814-21 [PubMed: 25250515]
  • Nava C, Hanna N, Michot C, Pereira S, Pouvreau N, Niihori T, Aoki Y, Matsubara Y, Arveiler B, Lacombe D, Pasmant E, Parfait B, Baumann C, Heron D, Sigaudy S, Toutain A, Rio M, Goldenberg A, Leheup B, Verloes A, Cave H (2007) CFC and Noonan syndromes due to mutations in RAS/MAPK signaling pathway: genotype/phenotype relationships and overlap with Costello syndrome. J Med Genet 44:763-71. [PMC free article: PMC2652823] [PubMed: 17704260]
  • Neumann TE, Allanson J, Kavamura I, Kerr B, Neri G, Noonan J, Cordeddu V, Gibson K, Tzschach A, Krüger G, Hoeltzenbein M, Goecke TO, Kehl HG, Albrecht B, Luczak K, Sasiadek MM, Musante L, Laurie R, Peters H, Tartaglia M, Zenker M, Kalscheuer V (2009) Multiple giant cell lesions in patients with Noonan syndrome and cardio-facio-cutaneous syndrome. Eur J Hum Genet. 17:420-5. [PMC free article: PMC2986220] [PubMed: 18854871]
  • Niemeyer CM, Kang MW, Shin DH, Furlan I, Erlacher M, Bunin NJ, Bunda S, Finklestein JZ, Sakamoto KM, Gorr TA, Mehta P, Schmid I, Kropshofer G, Corbacioglu S, Lang PJ, Klein C, Schlegel PG, Heinzmann A, Schneider M, Starý J, van den Heuvel-Eibrink MM, Hasle H, Locatelli F, Sakai D, Archambeault S, Chen L, Russell RC, Sybingco SS, Ohh M, Braun BS, Flotho C, Loh ML. Germline CBL mutations cause developmental abnormalities and predispose to juvenile myelomonocytic leukemia. Nat Genet. 2010;42:794-800. [PMC free article: PMC4297285] [PubMed: 20694012]
  • Niemeyer CM. RAS diseases in children. Haematologica. 2014;99:1653-62. [PMC free article: PMC4222471] [PubMed: 25420281]
  • Niihori T, Aoki Y, Ohashi H, Kurosawa K, Kondoh T, Ishikiriyama S, Kawame H, Kamasaki H, Yamanaka T, Takada F, Nishio K, Sakurai M, Tamai H, Nagashima T, Suzuki Y, Kure S, Fujii K, Imaizumi M, Matsubara Y (2005) Functional analysis of PTPN11/SHP-2 mutants identified in Noonan syndrome and childhood leukemia. J Hum Genet 50:192-202 [PubMed: 15834506]
  • Niihori T, Nagai K, Fujita A, Ohashi H, Okamoto N, Okada S, Harada A, Kihara H, Arbogast T, Funayama R, Shirota M, Nakayama K, Abe T, Inoue SI, Tsai IC, Matsumoto N, Davis EE, Katsanis N, Aoki Y. Germline-activating RRAS2 mutations cause Noonan syndrome. Am J Hum Genet. 2019;104:1233-40. [PMC free article: PMC6562005] [PubMed: 31130285]
  • Noonan JA (2001) Cardiac findings in cardio-facio-cutaneous syndrome: similarities to Noonan and Costello syndromes. Proc Greenwood Gen Ctr
  • Noonan JA. Noonan syndrome. In: Goldstein S, Reynolds CR, eds. Handbook of Neurodevelopmental and Genetic Disorders in Adults. New York: Guilford Press; 2005a:308-19.
  • Noonan Syndrome Guideline Development Group. Management of Noonan Syndrome – A Clinical Guideline. University of Manchester: DYSCERNE. Available online. 2010. Accessed 3-16-22.
  • Noordam C, Peer PG, Francois I, De Schepper J, van den Burgt I, Otten BJ. Long-term GH treatment improves adult height in children with Noonan syndrome with and without mutations in protein tyrosine phosphatase, non-receptor-type 11. Eur J Endocrinol. 2008;159:203-8. [PubMed: 18562489]
  • Noordam K (2007) Expanding the genetic spectrum of Noonan syndrome. Horm Res 68:24-7 [PubMed: 18174700]
  • Nyström A-M, Ekvall S, Berglund E, Björkvist M, Braathen G, Duchen K, Enell H, Holmberg E, Holmlund U, Olsson-Engman M, Annerén G, Bondeson M-L (2008) Noonan and cardio-facio-cutaneous syndromes: two clinically and genetically overlapping disorders. J Med Genet 45:500-6. [PubMed: 18456719]
  • Nyström AM, Ekvall S, Allanson J, Edeby C, Elinder M, Holmström G, Bondeson ML, Annerén G. (2009) Noonan syndrome and neurofibromatosis type I in a family with a novel mutation in NF1. Clin Genet. 76:524-34. [PubMed: 19845691]
  • Ogawa M, Moriya N, Ikeda H, Tanae A, Tanaka T, Ohyama K, Mori O, Yazawa T, Fujita K, Seino Y, Kubo T, Tanaka H, Nishi Y, Yoshimoto M (2004) Clinical evaluation of recombinant human growth hormone in Noonan syndrome. Endocr J 51:61-8 [PubMed: 15004410]
  • Osio D, Dahlgren J, Wikland KA, Westphal O (2005) Improved final height with long-term growth hormone treatment in Noonan syndrome. Acta Paediatr 94:1232-7 [PubMed: 16203673]
  • Pandit B, Sarkozy A, Pennacchio LA, Carta C, Oishi K, Martinelli S, Pogna EA, Schackwitz W, Ustaszewska A, Landstrom A, Bos JM, Ommen SR, Esposito G, Lepri F, Faul C, Mundel P, López Siguero JP, Tenconi R, Selicorni A, Rossi C, Mazzanti L, Torrente I, Marino B, Digilio MC, Zampino G, Ackerman MJ, Dallapiccola B, Tartaglia M, Gelb BD (2007) Gain-of-function RAF1 mutations cause Noonan and LEOPARD syndromes with hypertrophic cardiomyopathy. Nat Genet 39:1007-12 [PubMed: 17603483]
  • Pauta M, Martinez-Portilla R, Borrell A. Diagnostic yield of exome sequencing in fetuses with an isolated increased nuchal translucency: systematic review and meta-analysis. Ultrasound Obstet Gynecol. 2022;59:26-32. [PubMed: 34309942]
  • Pierpont EI. Neuropsychological functioning in individuals with Noonan syndrome: a systematic literature review with educational and treatment recommendations. J Pediatr Neuropsychol. 2016;2:14-33. Available online. Accessed 3-16-22.
  • Pierpont EI, Tworog-Dube E, Roberts AE. Attention skills and executive functioning in children with Noonan syndrome and their unaffected siblings. Dev Med Child Neurol. 2015;57:385-92. [PMC free article: PMC9979240] [PubMed: 25366258]
  • Pires LVL, Bordim RA, Maciel MBR, Tanaka ACS, Yamamoto GL, Honjo RS, Kim CA, Bertola DR. Atypical, severe hypertrophic cardiomyopathy in a newborn presenting Noonan syndrome harboring a recurrent heterozygous MRAS variant. Am J Med Genet A. 2021;185:3099-103. [PubMed: 34080768]
  • Prendiville TW, Gauvreau K, Tworog-Dube E, Patkin L, Kucherlapati RS, Roberts AE, Lacro RV. Cardiovascular disease in Noonan syndrome. Arch Dis Child. 2014;99:629-34. [PubMed: 24534818]
  • Quaio CR, de Almeida TF, Brasil AS, Pereira AC, Jorge AA, Malaquias AC, Kim CA, Bertola DR. Tegumentary manifestations of Noonan and Noonan-related syndromes. Clinics (Sao Paulo). 2013;68:1079-83. [PMC free article: PMC3752636] [PubMed: 24037001]
  • 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]
  • Roberts AE, Allanson JE, Tartaglia M, Gelb BD. Noonan syndrome. Lancet. 2013;381:333-42. [PMC free article: PMC4267483] [PubMed: 23312968]
  • Roberts AE, Araki T, Swanson KD, Montgomery KT, Schiripo TA, Joshi VA, Li L, Yassin Y, Tamburino AM, Neel BG, Kucherlapati RS (2007) Germline gain-of-function mutations in SOS1 cause Noonan syndrome. Nat Genet 39:70-4 [PubMed: 17143285]
  • Rodríguez FA, Castellón L, Moreno R, Paez E, Aracena Álvarez MI. Noonan syndrome with multiple Giant cell lesions, management and treatment with surgery and interferon alpha-2a therapy: case report. Birth Defects Res. 2020;112:732-9. [PubMed: 32065515]
  • Rohrer TR, Abuzzahab J, Backeljauw P, Birkegård AC, Blair J, Dahlgren J, Júlíusson PB, Ostrow V, Pietropoli A, Polak M, Romano A, Ross J, Sävendahl L, Miller BS. Long-Term Effectiveness and Safety of Childhood Growth Hormone Treatment in Noonan Syndrome. Horm Res Paediatr. 2020;93(6):380-395. [PubMed: 33440388]
  • Romano AA, Allanson JE, Dahlgren J, Gelb BD, Hall B, Pierpont ME, Roberts AE, Robinson W, Takemoto CM, Noonan JA. Noonan syndrome: clinical features, diagnosis, and management guidelines. Pediatrics. 2010;126:746-59. [PubMed: 20876176]
  • Romano AA, Dana K, Bakker B, Davis DA, Hunold JJ, Jacobs J, Lippe B. Growth response, near-adult height, and patterns of growth and puberty in patients with Noonan syndrome treated with growth hormone. J Clin Endocrinol Metab. 2009;94:2338-44. [PubMed: 19401366]
  • Runtuwene V, van Eekelen M, Overvoorde J, Rehmann H, Yntema HG, Nillesen WM, et al. Noonan syndrome gain-of-function mutations in NRAS cause zebrafish gastrulation defects. Dis Model Mech. 2011;4:393–9. [PMC free article: PMC3097460] [PubMed: 21263000]
  • Sana ME, Spitaleri A, Spiliotopoulos D, Pezzoli L, Preda L, Musco G, Ferrazzi P, Iascone M. Identification of a novel de novo deletion in RAF1 associated with biventricular hypertrophy in Noonan syndrome. Am J Med Genet A. 2014;164A:2069-73. [PubMed: 24782337]
  • Sarkozy A, Carta C, Moretti S, Zampino G, Digilio MC, Pantaleoni F, Scioletti AP, Esposito G, Cordeddu V, Lepri F, Petrangeli V, Dentici ML, Mancini GM, Selicorni A, Rossi C, Mazzanti L, Marino B, Ferrero GB, Silengo MC, Memo L, Stanzial F, Faravelli F, Stuppia L, Puxeddu E, Gelb BD, Dallapiccola B, Tartaglia M (2009) Germline BRAF mutations in Noonan, LEOPARD, and cardiofaciocutaneous syndromes: molecular diversity and associated phenotypic spectrum. Hum Mutat. 30:695-702. [PMC free article: PMC4028130] [PubMed: 19206169]
  • Schubbert S, Zenker M, Rowe SL, Böll S, Klein C, Bollag G, van der Burgt I, Musante L, Kalscheuer V, Wehner LE, Nguyen H, West B, Zhang KY, Sistermans E, Rauch A, Niemeyer CM, Shannon K, Kratz CP (2006) Germline KRAS mutations cause Noonan syndrome. Nat Genet 38:331-6 [PubMed: 16474405]
  • Seo GH, Yoo HW. Growth hormone therapy in patients with Noonan syndrome. Ann Pediatr Endocrinol Metab. 2018;23:176-81. [PMC free article: PMC6312920] [PubMed: 30599478]
  • Sharland M, Burch M, McKenna WM, Paton MA. A clinical study of Noonan syndrome. Arch Dis Child. 1992;67:178-83. [PMC free article: PMC1793396] [PubMed: 1543375]
  • Shchelochkov OA, Patel A, Weissenberger GM, Chinault AC, Wiszniewska J, Fernandes PH, Eng C, Kukolich MK, Sutton VR (2008) Duplication of chromosome band 12q24.11q24.23 results in apparent Noonan syndrome. Am J Med Genet A 146A:1042-8 [PubMed: 18348260]
  • Shaw AC, Kalidas K, Crosby AH, Jeffery S, Patton MA (2007) The natural history of Noonan syndrome: a long-term follow-up study. Arch Dis Child 92:128-32 [PMC free article: PMC2083343] [PubMed: 16990350]
  • Smpokou P, Tworog-Dube E, Kucherlapati RS, Roberts AE. Medical complications, clinical findings, and educational outcomes in adults with Noonan syndrome. Am J Med Genet A. 2012;158A:3106-11. [PubMed: 23165751]
  • Stevenson DA, Viskochil DH, Rope AF, Carey JC (2006) Clinical and molecular aspects of an informative family with neurofibromatosis type 1 and Noonan phenotype. Clin Genet. 69:246-53. [PMC free article: PMC3243644] [PubMed: 16542390]
  • Stuurman KE, Joosten M, van der Burgt I, Elting M, Yntema HG, Meijers-Heijboer H, Rinne T. Prenatal ultrasound findings of rasopathies in a cohort of 424 fetuses: update on genetic testing in the NGS era. J Med Genet. 2019;56:654-61. [PubMed: 31040167]
  • Suzuki H, Takenouchi T, Uehara T, Takasago S, Ihara S, Yoshihashi H, Kosaki K. Severe Noonan syndrome phenotype associated with a germline Q71R MRAS variant: a recurrent substitution in RAS homologs in various cancers. Am J Med Genet A. 2019;179:1628-30. [PubMed: 31173466]
  • Tartaglia M, Kalidas K, Shaw A, Song X, Musat DL, van der Burgt I, Brunner HG, Bertola DR, Crosby A, Ion A, Kucherlapati RS, Jeffery S, Patton MA, Gelb BD (2002) PTPN11 mutations in Noonan syndrome: molecular spectrum, genotype-phenotype correlation, and phenotypic heterogeneity. Am J Hum Genet 70:1555-63 [PMC free article: PMC379142] [PubMed: 11992261]
  • Tartaglia M, Martinelli S, Cazzaniga G, Cordeddu V, Iavarone I, Spinelli M, Palmi C, Carta C, Pession A, Aricò M, Masera G, Basso G, Sorcini M, Gelb BD, Biondi A (2004) Genetic evidence for lineage-related and differentiation stage-related contribution of somatic PTPN11 mutations to leukemogenesis in childhood acute leukemia. Blood 104:307-13 [PubMed: 14982869]
  • Tartaglia M, Niemeyer CM, Fragale A, Song X, Buechner J, Jung A, Hählen K, Hasle H, Licht JD, Gelb BD (2003) Somatic mutations in PTPN11 in juvenile myelomonocytic leukemia, myelodysplastic syndromes and acute myeloid leukemia. Nat Genet 34:148-50 [PubMed: 12717436]
  • Tartaglia M, Pennacchio LA, Zhao C, Yadav KK, Fodale V, Sarkozy A, Pandit B, Oishi K, Martinelli S, Schackwitz W, Ustaszewska A, Martin J, Bristow J, Carta C, Lepri F, Neri C, Vasta I, Gibson K, Curry CJ, Siguero JP, Digilio MC, Zampino G, Dallapiccola B, Bar-Sagi D, Gelb BD (2007) Gain-of-function SOS1 mutations cause a distinctive form of Noonan syndrome. Nat Genet 39:75-9 [PubMed: 17143282]
  • Tofil NM, Winkler MK, Watts RG, Noonan J (2005) The use of recombinant factor VIIa in a patient with Noonan syndrome and life-threatening bleeding. Pediatr Crit Care Med 6:352-4 [PubMed: 15857538]
  • van der Burgt I (2007) Noonan syndrome. Orphanet J Rare Dis 2:4. [PMC free article: PMC1781428] [PubMed: 17222357]
  • van der Burgt I, Thoonen G, Roosenboom N, Assman-Hulsmans C, Gabreels F, Otten B, Brunner HG (1999) Patterns of cognitive functioning in school-aged children with Noonan syndrome associated with variability in phenotypic expression. J Pediatr 135:707-13 [PubMed: 10586173]
  • van Nierop JWI, van Trier DC, van der Burgt I, Draaisma JMT, Mylanus EAM, Snik AF, Admiraal RJC, Kunst HPM. Cochlear implantation and clinical features in patients with Noonan syndrome and Noonan syndrome with multiple lentigines caused by a mutation in PTPN11. Int J Pediatr Otorhinolaryngol. 2017;97:228-34. [PubMed: 28483241]
  • van Trier DC, van der Burgt I, Draaijer RW, Cruysberg JRM, Noordam C, Draaisma JM. Ocular findings in Noonan syndrome: a retrospective cohort study of 105 patients. Eur J Pediatr. 2018 Aug;177(8):1293-1298. [PMC free article: PMC6061046] [PubMed: 29948256]
  • Villani A, Greer MC, Kalish JM, Nakagawara A, Nathanson KL, Pajtler KW, Pfister SM, Walsh MF, Wasserman JD, Zelley K, Kratz CP. Recommendations for cancer surveillance in individuals with RASopathies and other rare genetic conditions with increased cancer risk. Clin Cancer Res. 2017;23:e83-e90. [PubMed: 28620009]
  • Vissers LE, Bonetti M, Paardekooper Overman J, Nillesen WM, Frints SG, de Ligt J, Zampino G, Justino A, Machado JC, Schepens M, Brunner HG, Veltman JA, Scheffer H, Gros P, Costa JL, Tartaglia M, van der Burgt I, Yntema HG, den Hertog J. Heterozygous germline mutations in A2ML1 are associated with a disorder clinically related to Noonan syndrome. Eur J Hum Genet. 2015;23:317-24. [PMC free article: PMC4326711] [PubMed: 24939586]
  • Wilkinson JD, Lowe AM, Salbert BA, Sleeper LA, Colan SD, Cox GF, Towbin JA, Connuck DM, Messere JE, Lipshultz SE. Outcomes in children with Noonan syndrome and hypertrophic cardiomyopathy: a study from the Pediatric Cardiomyopathy Registry. Am Heart J 2012;164:442-8 [PubMed: 22980313]
  • Wolvius EB, de Lange J, Smeets EE, van der Wal KG, van den Akker HP (2006) Noonan-like/multiple giant cell lesion syndrome: report of a case and review of the literature. J Oral Maxillofac Surg 64:1289-92 [PubMed: 16860226]
  • Yamamoto GL, Aguena M, Gos M, Hung C, Pilch J, Fahiminiya S, Abramowicz A, Cristian I, Buscarilli M, Naslavsky MS, Malaquias AC, Zatz M, Bodamer O, Majewski J, Jorge AA, Pereira AC, Kim CA, Passos-Bueno MR, Bertola DR. Rare variants in SOS2 and LZTR1 are associated with Noonan syndrome. J Med Genet. 2015;52:413-21. [PubMed: 25795793]
  • Yaoita M, Niihori T, Mizuno S, Okamoto N, Hayashi S, Watanabe A, Yokozawa M, Suzumura H, Nakahara A, Nakano Y, Hokosaki T, Ohmori A, Sawada H, Migita O, Mima A, Lapunzina P, Santos-Simarro F, García-Miñaúr S, Ogata T, Kawame H, Kurosawa K, Ohashi H, Inoue S, Matsubara Y, Kure S, Aoki Y. Spectrum of mutations and genotype-phenotype analysis in Noonan syndrome patients with RIT1 mutations. Hum Genet. 2016;135:209-22. [PubMed: 26714497]
  • Yoon SR, Choi SK, Eboreime J, Gelb BD, Calabrese P, Arnheim N. Age-dependent germline mosaicism of the most common Noonan syndrome mutation shows the signature of germline selection. Am J Hum Genet. 2013;92:917-26. [PMC free article: PMC3682763] [PubMed: 23726368]
  • Yoshida R, Miyata M, Nagai T, Yamazaki T, Ogata T (2004) A 3-bp deletion mutation of PTPN11 in an infant with severe Noonan syndrome including hydrops fetalis and juvenile myelomonocytic leukemia. Am J Med Genet A. 128A:63-6. [PubMed: 15211660]
  • Zarate YA, Lichty AW, Champion KJ, Clarkson LK, Holden KR, Matheus MG. Unique cerebrovascular anomalies in Noonan syndrome with RAF1 mutation. J Child Neurol. 2014;29:NP13-7. [PubMed: 23877478]
  • Zavras N, Meazza C, Pilotta A, Gertosio C, Pagani S, Tinelli C, Bozzola M. Five-year response to growth hormone in children with Noonan syndrome and growth hormone deficiency. Ital J Pediatr. 2015;41:71. [PMC free article: PMC4595189] [PubMed: 26444854]
  • Zenker M, Lehmann K, Schulz AL, Barth H, Hansmann D, Koenig R, Korinthenberg R, Kreiss-Nachtsheim M, Meinecke P, Morlot S, Mundlos S, Quante AS, Raskin S, Schnabel D, Wehner LE, Kratz CP, Horn D, Kutsche K (2007) Expansion of the genotypic and phenotypic spectrum in patients with KRAS germline mutations. J Med Genet 44:131-5 [PMC free article: PMC2598066] [PubMed: 17056636]
Copyright © 1993-2024, University of Washington, Seattle. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved.

GeneReviews® chapters are owned by the University of Washington. Permission is hereby granted to reproduce, distribute, and translate copies of content materials for noncommercial research purposes only, provided that (i) credit for source (http://www.genereviews.org/) and copyright (© 1993-2024 University of Washington) are included with each copy; (ii) a link to the original material is provided whenever the material is published elsewhere on the Web; and (iii) reproducers, distributors, and/or translators comply with the GeneReviews® Copyright Notice and Usage Disclaimer. No further modifications are allowed. For clarity, excerpts of GeneReviews chapters for use in lab reports and clinic notes are a permitted use.

For more information, see the GeneReviews® Copyright Notice and Usage Disclaimer.

For questions regarding permissions or whether a specified use is allowed, contact: ude.wu@tssamda.

Bookshelf ID: NBK1124PMID: 20301303

Views

Related information

  • MedGen
    Related information in MedGen
  • OMIM
    Related OMIM records
  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed
  • Gene
    Locus Links

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...