Next Article in Journal
Neuroimaging in Adolescents: Post-Traumatic Stress Disorder and Risk for Substance Use Disorders
Next Article in Special Issue
NUP85 as a Neurodevelopmental Gene: From Podocyte to Neuron
Previous Article in Journal
Physiological, Photosynthetic, and Transcriptomics Insights into the Influence of Shading on Leafy Sweet Potato
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Cardiofaciocutaneous Syndrome: From Genetics to Prognostic–Therapeutic Implications

by
Giovanna Scorrano
1,
Emanuele David
2,
Elisa Calì
3,
Roberto Chimenz
4,
Saverio La Bella
1,
Armando Di Ludovico
1,
Gabriella Di Rosa
5,
Eloisa Gitto
6,
Kshitij Mankad
7,
Rosaria Nardello
8,
Giuseppe Donato Mangano
8,
Chiara Leoni
9,* and
Giorgia Ceravolo
3
1
Department of Pediatrics, “G. D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
2
Department of Translational and Precision Medicine, “Sapienza” University of Rome, 00161 Rome, Italy
3
UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
4
Pediatric Nephrology and Dialysis Unit, University Hospital “G. Martino”, 98124 Messina, Italy
5
Child Neuropsychiatry Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98124 Messina, Italy
6
Neonatal and Pediatric Intensive Care Unit, Department of Human Pathology of the Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98122 Messina, Italy
7
Department of Radiology, Great Ormond Street Hospital for Children, London WC1N 3JH, UK
8
Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialities, “G. D’Alessandro” University of Palermo, 90127 Palermo, Italy
9
Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
*
Author to whom correspondence should be addressed.
Genes 2023, 14(12), 2111; https://doi.org/10.3390/genes14122111
Submission received: 9 October 2023 / Revised: 16 November 2023 / Accepted: 21 November 2023 / Published: 22 November 2023
(This article belongs to the Special Issue Genetics in Pediatric Neurodevelopmental Disorders)

Abstract

:
Cardiofaciocutaneous (CFC) syndrome is one of the rarest RASopathies characterized by multiple congenital ectodermal, cardiac and craniofacial abnormalities with a mild to severe ocular, gastrointestinal and neurological involvement. It is an autosomal dominant syndrome, with complete penetrance, caused by heterozygous pathogenic variants in the genes BRAF, MAP2K1/MEK1, MAP2K2/MEK2, KRAS or, rarely, YWHAZ, all part of the RAS-MAPK pathway. This pathway is a signal transduction cascade that plays a crucial role in normal cellular processes such as cell growth, proliferation, differentiation, survival, metabolism and migration. CFC syndrome overlaps with Noonan syndrome, Costello syndrome, neurofibromatosis type 1 and Legius syndrome, therefore making the diagnosis challenging. Neurological involvement in CFC is more severe than in other RASopathies. Phenotypic variability in CFC patients is related to the specific gene affected, without a recognized genotype–phenotype correlation for distinct pathogenic variants. Currently, there is no specific treatment for CFC syndrome. Encouraging zebrafish model system studies suggested that, in the future, MEK inhibitors could be a suitable treatment of progressive phenotypes of CFC in children. A multidisciplinary care is necessary for appropriate medical management.

1. Introduction

CFC syndrome (OMIM 115150) is a multisystemic disorder that affects 1 in 800,000 newborns. CFC belongs to a group of syndromes collectively known as “RASopathies”, caused by germline mutations in one of the genes encoding components of the RAS-MAPK signal transduction pathway [1,2]. The RAS-MAPK pathway is a signal transduction cascade that plays a crucial role in normal cellular processes such as cell growth, proliferation, differentiation, survival, metabolism and migration. Somatic gain-of-function mutations in RAS genes are a well-known cause of cancer and there has been an increasing number of studies involving the RAS pathway in oncogenesis [3]. Though each syndrome has its specific phenotype, many of the RASopathies have overlapping clinical features, therefore making the diagnosis challenging. These can include facial dysmorphisms, intellectual disability (ID), congenital heart disease, short stature, muscle skeletal anomalies and lymphatic dysfunction. CFC syndrome may clinically overlap with neurofibromatosis type 1 (NF1), Noonan syndrome (NS), Noonan syndrome with multiple lentigines (NS-ML), Costello syndrome (CS), Legius syndrome (LS), central conducting lymphatic anomalies syndrome (CCLA), SYNGAP1 syndrome, and capillary malformation arteriovenous malformation syndrome (CM-AVM), though compared with other RASopathies, it exhibits more severe neurologic complications and phenotype [4]. CFC patients frequently present with a characteristic facial appearance, congenital heart disease, severe developmental delay and psychomotor delay, ectodermal, ocular and gastrointestinal abnormalities, and neurological involvement [1]. Characteristic facial features include high forehead, bitemporal constriction, supraorbital hypoplasia, downslanting palpebral fissures, depressed nasal bridge and posteriorly rotated ears with thick helices [5,6]. Compared to other RASopathies, neurological involvement is more severe and commonly includes refractory epilepsy, neurocognitive impairment, motor and speech deficits, hypotonia and behavioral challenges; ventriculomegaly, cortical atrophy and hydrocephalus are the most frequent imaging findings in CFC patients [6,7]. Cardiac involvement is common in CFC and manifests as atrial and ventricular septal defects, hypertrophic cardiomyopathy and pulmonary valve stenosis. These patients also have curly and friable hair, and sparse eyebrows and eyelashes; skin involvement ranges from dry skin to hyperkeratosis. CFC is inherited in an autosomal-dominant manner, and most pathogenic variants involve the genes BRAF (75%), MAP2K1 (25%), MAP2K2 (25%), KRAS (2%) or, more rarely, YWHAZ [8]. The real incidence is still unknown and a phenotypic variability related to the specific gene affected has been identified, without a genotype–phenotype correlation for specific pathogenic variants (Table 1) [5]. The diagnosis of CFC syndrome is suspected based on the distinctive clinical features and it is confirmed by molecular genetic testing [5,9]. The aim of the study is to give updated evidence on the pathogenesis, clinical and radiological features, diagnosis, and available treatment for CFC, supplying practice guidelines for the management of pediatric patients with CFC.

2. Methodology

In this narrative review, we searched articles on PubMed up to September 2023. The search formula was as follows: (((CFC) OR (Cardiofaciocutaneous Syndrome)) AND ((gene pathogenic variants) OR (mutations) OR (related neurological disorders) OR (neuroradiological findings) OR (related skin anomalies) OR (related gastrointestinal disorders) OR (muscle-skeletal findings) OR (behavior and cognitive disorders) OR (cardiac disorders) OR (craniofacial findings) OR (endocrinologic and growth disorders))).
We selected studies, published in English, that included CFC pathogenesis, clinical and radiological features, diagnosis, and the latest evidence on new treatments.
We revised the literature, exploring the evolving spectrum related to pathogenic variants affecting patients with CFC, described to date. Of note, we firstly reported systematically the phenotypic variability related to the specific gene affected in the syndrome, with relevant prognostic–therapeutic implications. Additionally, we first mentioned in the review the new gene causally related to CFC, YWHAZ, describing how de novo missense pathogenic variants in this gene lead to RAS/MAPK pathway disruption and clinical manifestations. The family involved in the study provided informed consent for being part of this study.

3. Pathogenesis

Currently, BRAF, MAP2K1, MAP2K2 and, rarely, KRAS have been associated with CFC encoding components of the RAS-MAPK pathway. The signal cascade of RAS-MAPK is highly conserved and plays a crucial role in normal cellular processes as cell growth, proliferation, differentiation, survival, metabolism and migration [5,10]. The pathway is stimulated by growth factors that lead RAS-mediated RAF activation. This kinase phosphorylates MEK1 and/or MEK2 with consequent ERK1/2 activation, downstream the cascade [11]. ERK1 and ERK2 effectors act both in the nucleus and in the cytoplasm, mediating the polymorphic cellular response to growth factors (Figure 1).
Pathogenic variants are mainly missense with a gain-of-function mechanism on proteins of the pathway, leading to ERK1-2 hyperactivation.
The RAS-MAPK pathway is a signal transduction cascade that plays a crucial role in normal cellular processes such as cell growth, proliferation, differentiation, survival, metabolism and migration. The pathway is stimulated by growth factors that lead to RAS-mediated RAF activation. This kinase phosphorylates MEK1 and/or MEK2 with consequent ERK1/2 activation, downstream the cascade. ERK1 and ERK2 effectors act both in nucleus and in cytoplasm, mediating the polymorphic cellular response to growth factors.

4. Neurological Findings

Neurological involvement in CFC syndrome is present in nearly all individuals and ranges from mild to severe. Developmental delay, ID, epilepsy and hypotonia are common findings, although a few individuals may have normal cognitive function (IQ within normal range).
In children with CFC, seizures frequently occurred, usually with onset in infancy or early childhood and they included tonic–clonic and/or complex focal seizures, absence seizures and epileptic infantile spasms [7,12,13,14,15,16]; seizures could occur later in infancy, requiring polytherapy and refractoriness to antiseizure medications (ASMs) [5]. However, in children, seizure may occur also due to multiple different genetic conditions [17,18,19,20].
Hypotonia is also described and presented typically with a neonatal onset with mild to severe asthenia, motor delay and poor muscle mass [5]. Furthermore, behavior abnormalities could characterize a neuropsychiatric disorder in CFC patients, including irritability, obsessive compulsive disorder, anxiety and autistic traits [5,21].
A recent clinical study by Pierpont et al. [6] reported a higher rate of CFC patients affected by epilepsy than in the previously published literature: seizures of various type occurred in 57% of individuals with BRAF pathogenic variants, 61% of patients with MAP2K1 pathogenic variants and 30% of subjects with MAP2K2 pathogenic variants. MAP2K2 mutations were associated with a lower risk of seizure occurrence and less severe seizure types [6]. On the other hand, patients with pathogenic variants in BRAF and MAP2K1 presented with moderate to severe polymorphic seizures, which were frequently drug-resistant. Furthermore, a relevant study by Battaglia et al. [16] shed a light on genotype–phenotype correlation concerning the epileptic phenotype. There seems to be a strong genotype–phenotype correlation regarding the occurrence and severity of seizures, since variants in the protein kinase domain of BRAF (exons 11–16) and the p.Y130C/H/N variant of MAP2K1 (exon 3) correlate with a severe epileptic phenotype [6]. Nevertheless, pathogenic variants in KRAS variants were not associated with epilepsy. Notably, most children affected with the above genotypes and severe epilepsy were also presenting neurodevelopmental impairment. ID was reported in most individuals (82 -100%), usually carrying BRAF (89%) or MAP2K1 (84%) pathogenic variants, while it was described only in 25% of patients with MAP2K2 pathogenic variants [6]. The degree of ID ranged from mild (MEK) to moderate (BRAF) [22]. Neurodevelopmental delay may be mild and difficult to notice in patients with mild or moderate involvement, while speech and motor delays are usually obvious in severely affected children [23]. Nevertheless, those are non-specific findings, often found also in children with other kinds of neurologic impairment [19,24,25,26]. Motor delay and hypotonia are the most frequently observed neurological features in CFC syndrome [27]. Pierpont et al., reported a distinctive genotype–phenotype correlation: children carrying pathogenic variants in MAP2K1 showed more need for support and lack of independent ambulation (71%) compared to patients with BRAF (29%) or MAP2K2 (13%) mutations [6]. Language acquisition is usually delayed and speech abilities range from full sentences to non-verbal communications (9–31%) [9]. Interestingly, mutations promoting dysregulation of the RAS-MAPK cascade have been associated with an increased psychopathological risk. Frequently, patients with CFC presented underdiagnosed autistic-like behaviors [21].
A magnetic resonance imaging (MRI) evaluation could be useful during the diagnostic work-up. Brain magnetic resonance imaging studies may prove useful in patients with uncertain diagnosis and for prognostic information. Ventriculomegaly (43.9%) is the most common finding and sometimes requires a ventriculo-peritoneal shunt [28,29].
Other findings comprise prominent Virchow–Robin spaces (10.6%), hydrocephalus (24.2%) and myelination abnormalities. Some patients have structural brain anomalies, including Type I Chiari malformation (4.5%), arachnoid cyst (1.5%) and subependymal grey matter heterotopia (7.5%) [12].
Other abnormalities can include abnormal EEG, dilated perivascular spaces, dilated perivascular spaces, corticospinal tract findings, agenesis of the corpus callosum (6%), frontal lobe hypoplasia (1.5%), pachygyria (7.5%) and Chiari malformation (4.5%) [5] (Figure 2).

4.1. Cardiac Findings

Congenital heart defects (CHDs) occur in most people with CFC syndrome (~75%) [9,30,31,32]. The cardiac defects most frequently associated with this condition include pulmonary valvular stenosis (PVS) (~45%), Hypertrophic cardiomyopathy (HCM) (~40%), atrial septal defect (18–28%), ventricular septal defect (11–22%) and other anomalies such as mitral valve dysplasia, arrhythmias, tricuspid valve dysplasia, and bicuspid aortic valve [9]. Patients with MAP2K1 variants have a lower frequency of cardiac disease than those with MAP2K2 (64%) and BRAF variants (72%) [8]. Literature data show that neurological and cardiac phenotypes do not segregate in a similar pattern, so different variants in CFC genes have different consequences across tissues [6].

4.2. Ectodermal Findings

Ectodermal anomalies are cardinal features of CFC syndrome; virtually all patients with CFC syndrome develop some kind of ectodermal involvement.
The most common manifestations are sparse, curly, fine, brittle slow-growing hair; sparse to absent eyebrows with ulerythema ophryogenes; sparse to absent eyelashes; dystrophic rapid-growing nails; skin abnormalities such as keratosis pilaris, hyperkeratosis, ichthyosis, eczemas, xerosis, hemangiomas and numerous pigmented naevi (more common in patients with BRAF mutations) [33,34,35,36]. Some skin anomalies evolve with age: xerosis and follicular hyperkeratosis can improve [37], palmoplantar hyperkeratosis tends to worsen, especially in pressure areas, lymphedema may become more severe, and pigmented naevi [38,39] tend to grow in number [40]. It is also common for CFC patients to be affected by severe skin infections [5].

4.3. Craniofacial Findings

Children with CFC syndrome have characteristic craniofacial features. Those include macrocephaly, high forehead, bitemporal constriction, hypoplasia of the supraorbital ridges, downslanting palpebral fissures (more frequent in patients with MEK mutations than BRAF), ocular hypertelorism (more commonly associated with mutations in BRAF), ptosis, epicanthal folds, telecanthus, short nose with depressed bridge and anteverted nares, low-set and posteriorly angulated ears with prominent helices, high-arched palate and relative micrognathia [9].

4.4. Gastrointestinal and Growth Findings

Feeding problems are very frequently reported, almost in all CFC patients, and there does not seem to be any genotype–phenotype correlation [41,42]. These problems are usually reported in the neonatal period and manifest as poor suck, swallow dysfunction, vomiting, gastroesophageal reflux, aspiration, and oral aversion. Feeding issues are usually followed by inadequate oral caloric intake, failure to thrive, and often require nasogastric tube feeding (40%) or gastrotomy tube placement (50%) [9]. In these children, growth is usually delayed with both weight and length below normal range [5]. Oral aversion can persist during childhood but usually GI symptoms improve with age, other than in patients with epilepsy onset in childhood. Their GI disorders become worse with age. Constipation is often seen in children with CFC, other findings include intestinal malrotation, hernia and also splenomegaly, hepatomegaly and steatosis. Many common symptoms are a consequence of dysmotility including gastroesophageal reflux, vomiting and constipation (Figure 3) [22].
It is important to notice that, though the short stature could be related to a growth delay caused by poor feeding, it could also be linked to a growth hormone deficiency, since the RAS-MAPK pathway plays an important role in the insulin-like growth factor I (IGF-I) mediations and growth hormone (GH) secretion.

4.5. Additional Features

During perinatal period polyhydramnios (77%) and prematurity (up to 50%) have been reported as common findings [9,22].
The vast majority of individuals present with musculoskeletal abnormalities including hypotonia, pectus excavatum and carinatum, scoliosis, kyphosis, short neck and pterygium coli. The orthopedic features are more common in patients with MAP2K1 variant than in patients with other variants according to Leoni et al. [43]. Interestingly, patients with CFC present a reduced bone mineral density (BMD), probably associated with reduced physical activity and inflammatory cytokines. Despite vitamin D supplementation and almost normal bone metabolism biomarkers, CFC patients showed significantly decreased absolute values of DXA-assessed subtotal and lumbar BMD (p ≤ 0.05), compared to controls, with BMD z-scores and t-scores below the reference range in CFC, and normal in healthy controls [44].
Endocrine complications have been evaluated in patients with CFC. Specifically, a high prevalence of thyroid autoimmunity, with an increased risk to develop autoimmune disorders, and short stature have been described and presumably related to the dysregulation of the RAS-MAPK pathway, the reduced physical activity, the presence of inflammatory cytokines and the impaired IGF1 activity [45].
Ocular manifestations including strabismus, nystagmus, ocular hypertelorism, astigmatism, optic nerve hypoplasia, myopia, hyperopia, cortical blindness, and cataracts are present in most individuals with CFC syndrome and may result in poor visual acuity. Optic nerve hypoplasia is more commonly reported in patients with BRAF mutations [22,46].
Notably, functional limitations, pain and disability have been reported in patients with CFC. Using the Pediatric Outcomes Data Collection Instrument (PODCI) and Pediatric Evaluation of Disability Index (PEDI), an interesting study [27] documented that CFC patients presented activity limitations in the PODCI domains of upper extremity function, transfers and mobility, sport, and physical function. Concurrently, a relevant disability in the PEDI domains of daily activity, mobility, socialization and cognition was noted. Pain is highly prevalent in patients with RASopathies and CFC, as well. Specifically, musculoskeletal and abdominal pain was more frequently reported and often interfered with daily activities. Furthermore, pain negatively impacted QoL and sleeping patterns [47].
Interestingly, neoplasms, commonly observed in other RASopathies, have not been reported in CFC syndrome, even though genetic mutations causing CFC syndrome are well known to play a crucial role in development and oncogenesis. BRAF mutations have been associated with 7% of cancers [48,49], and there are few published reports of neoplasia in CFC [23,50], with only one malignancy [51,52].
Urogenital anomalies may occur in ~33% of individuals with CFC: cryptorchidism is the most common feature (66% of affected males); also, renal cysts, nephrolithiasis and bladder abnormalities may be present.

5. Diagnosis

The diagnosis of CFC syndrome is often challenging, and it should be based on a specific clinical manifestation and confirmed through molecular genetic testing.
The most common clinical features are short stature, distinctive craniofacial features, developmental delay, neuromotor delay, ID, ectodermal abnormalities, congenital heart disease, musculoskeletal features, feeding and gastrointestinal problems, and cryptorchidism. Features of CFC syndrome overlap considerably with some other RASopathies, including Costello syndrome, and Noonan syndrome. Some features may be present since birth while others may appear later in life; also, these clinical conditions can change over time either by becoming better or becoming more severe. Phenotypic variability in CFC is also associated with different variants in CFC genes. These phenotypic changes, overlaps and variability create diagnostic difficulties [6].
CFC syndrome is an autosomal-dominant syndrome, with complete penetrance [5,23].
An affected person has a 50% chance of passing the altered gene to each child. The mutation genes involved in CFC syndrome are BRAF (7q34) in up to 75% of the cases, MAP2K1 (15q22.1-q22.33) and MAP2K2 (19p13.3), both around 25% of the cases, and KRAS (12p12.1) present in less than 2% of the cases. Most individuals with CFC syndrome reported to date have a de novo BRAF, MAP2K1, MAP2K2, or KRAS pathogenic variant. Even though CFC is an autosomal dominant disorder, sporadic cases are the majority. To date, only seven families were reported as characterized by a vertical transmission of MEK2, KRAS, and BRAF pathogenic variants, respectively. This suggests that CFC pathogenic variants within the Ras/MAPK pathway are compatible with human reproduction. Additionally, the reproductive success may be affected by CFC phenotypic features, such as neurodevelopmental delay, instead of the disruption of the Ras/MAPK pathway. An intrafamilial variable expressivity has been identified, as well [53].
Recently, YWHAZ, a 14-3-3 family member, has been identified as a new gene involved in CFC. It was documented that the S230W YWHAZ variant enhanced Raf-stimulated Erk phosphorylation to a higher level than wild type, with a gain-of-function mechanism in the RAS-ERK pathway. This result supported that the variant was the underlying cause of the CFC phenotype [8]. Mutations in these genes cause a dysregulation of the RAS/MAPK signaling pathway. This pathway is critically involved in cell differentiation, proliferation, migration, and apoptosis, and is one of the most studied signaling cascades.
The genetic diagnosis of CFC syndrome is established by the identification of a heterozygous pathogenic variant in BRAF, MAP2K1, MAP2K2, or KRAS genes.

Current Consensus Guidelines Strategy Include the Following

  • Multigene panel for common RASopathy genes that includes BRAF, MAP2K1, MAP2K2, KRAS and YWHAZ usually detects up to 90% of individuals with CFC and it is the preferred initial test.
  • Individual single-gene testing is recommended if panel testing is not available, beginning with BRAF, MAP2K1, and MAP2K2, and KRAS.
  • If these molecular genetic tests are negative, a more comprehensive genetic sequencing including exome and genome sequencing should be performed [5].
In the event that a pathogenic variant is identified, prenatal testing should be recommended to determine if the mutations is inherited [5,54].

6. Treatment

Currently, there is no specific treatment in CFC syndrome. More recently, genetic pathway inhibitors, including MEK inhibitors, led to size reduction of inoperable plexiform neurofibromas (PNs) in NF1, with fewer side effects [55]. Moreover, in several mouse or other model systems, MEK inhibitors were used as a prenatal preventative therapy or postnatal treatment of non-NF1 RASopathies. In this context, in the in vivo zebrafish model system, it was observed that CFC mutations in BRAF and MEK impaired convergence–extension cell movements during gastrulation and MEK inhibitors prevented the cell migration defects caused by this pathogenetic variants in CFC, treating the embryos within a specific developmental time-window [11]. Additionally, an interesting functional study using human dermal lymphatic endothelial cells (HDLECs) and zebrafish larvae, to model KRAS pathogenic variants, documented an increased ERK phosphorylation with biochemical and morphological changes, significantly rescued by the subsequent use of MEK inhibitors [56]. Interestingly, two studies revealed how MEK inhibitors represented a promising treatment in HCM related to CFC syndrome, especially if administered before the onset of irreversible cardiac remodeling [57,58].
Moreover, a trial focusing on use of a MEK1/2 inhibitor, Trametinib, in NF1 or GNA11/Q alterations, did not demonstrate meaningful clinical activity. However, it appeared effective in one patient with multiple cancers, warranting future studies [59]. Furthermore, it was observed that Braf+/+ and BrafQ241R/+ mice treated with C-type natriuretic peptide (CNP), a stimulator of endochondral bone growth and a potent inhibitor of the FGFR3-RAF1-MEK/ERK signaling, presented increased body and tail lengths, suggesting that CNP could be a potential therapeutic target in CFC syndrome [60]. Additionally, BrafQ241R/+ knock-in mice showing cardiovascular and lymphatic defects ameliorated after combination treatment with a MEK inhibitor, PD0325901, and a histone 3 demethylase inhibitor, GSK-J4, suggesting that epigenetic modulation as well as the inhibition of the ERK pathway could be a potential therapeutic strategy [61].
These results suggest that small molecule inhibitors could be potentially used to treat the progressive phenotypes of CFC in children. Hence, future pre-clinical models of CFC should be developed, to better define protein interactions of the RAS/MAPK pathway, the pathophysiology of the disorder, and endpoints for measuring treatment efficacy [62].
Multidisciplinary care is crucial in CFC children; despite this, there is no specific treatment. Specifically, a comprehensive management should be performed, especially for individuals at risk. If patients are at risk for pulmonary stenosis, HCM and other cardiac defects, they should be submitted to a cardiac follow-up with blood pressure measurement at each visit and echocardiogram every 2–3 y up to 20 years of age, if no cardiac disease is found initially. Concurrently, frequent dermatology visits for the management of skin abnormalities such as keratosis pilaris, hyperkeratosis, ichthyosis, eczemas, xerosis, hemangiomas and annual evaluation of pigmented naevi should be performed. Furthermore, individuals at risk for infantile spasms, seizures, and brain anomalies should be evaluated from neurologists in a continuous follow-up. Functional behavior assessment, special education services, and early childhood intervention programs should be considered in patients with cognitive and behavior disorders, as well. If gastrointestinal difficulties are present, a regular follow-up to monitor growth and nutrition should be performed, with the measurement of growth parameters and nutritional status at each visit. Moreover, ophthalmologic defects should be controlled every six to 12 months, whereas audiologic evaluation every two to three years. Concurrently, patients with musculoskeletal anomalies, genitourinary malformations, hematologic disorders and dental lesions should be systematically followed by specialists to prevent complications, improving the quality of life of these complex patients [5,9].

7. Conclusions

In recent years, studies based on NGS and omics-related sciences revealed an expanding molecular complexity underlying genetic disorders such as RASopathies [63,64,65,66,67]. Many novel molecular factors and genes have been identified with consequent benefits in terms of refining clinical phenotypes, valuable prognostic information, detailed imaging studies and targeted therapies for the children affected with these conditions [68,69,70,71,72]. The CFC phenotype is very heterogeneous, as it depends on the gene involved, as well as the type and localization of the pathogenic variant, and it can change and evolve across time. Like other RASopathies, CFC syndrome presents distinctive clinical features and involves numerous organ systems. Even though, there is no specific treatment in CFC syndrome, encouraging zebrafish model system studies suggested that MEK inhibitors could be a suitable targeted treatment of progressive phenotypes of CFC in future. Currently, a multidisciplinary care is necessary to manage the numerous issues that are present in this condition: neurological, cardiological, gastroenterological, endocrinological, dermatological, orthopedic, ophthalmological, and behavioral problems. Recent clinical studies highlighted the strong and significant correlations between different phenotypes and gene variants. Investigations to better understand the disease pathway in this pediatric population is needed and it will hopefully enable and guide more effective therapeutic avenues in the future.

Author Contributions

G.S., C.L. and E.D. contributed to the conception of review and structure. G.C. and G.S. performed the literature review and wrote the first draft of the manuscript. C.L., R.C., S.L.B., A.D.L., E.G., R.N., K.M., G.D.M., G.D.R. and E.C. contributed additional literature sources for review and content revision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

The family involved in the study provided informed consent for being part of this study.

Data Availability Statement

Data are contained within the article.

Acknowledgments

We want to thank the Italian Association of Costello and Cardiofaciocutaneous Syndrome—RASopathies (AISC).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hebron, K.E.; Hernandez, E.R.; Yohe, M.E. The RASopathies: From pathogenetics to therapeutics. Dis. Models Mech. 2022, 15, dmm049107. [Google Scholar] [CrossRef] [PubMed]
  2. Abe, Y.; Aoki, Y.; Kuriyama, S.; Kawame, H.; Okamoto, N.; Kurosawa, K.; Ohashi, H.; Mizuno, S.; Ogata, T.; Kure, S.; et al. Prevalence and clinical features of Costello syndrome and cardio-facio-cutaneous syndrome in Japan: Findings from a nationwide epidemiological survey. Am. J. Med. Genet. Part A 2012, 158, 1083–1094. [Google Scholar] [CrossRef] [PubMed]
  3. Lim, J.K.M.; Leprivier, G. The impact of oncogenic RAS on redox balance and implications for cancer development. Cell Death Dis. 2019, 10, 955. [Google Scholar] [CrossRef]
  4. Zenker, M. Clinical overview on RASopathies. Am. J. Med. Genet. C Semin. Med. Genet. 2022, 190, 414–424. [Google Scholar] [CrossRef]
  5. Rauen, K.A. Cardiofaciocutaneous Syndrome. In GeneReviews®; Adam, M.P., Mirzaa, G.M., Pagon, R.A., Wallace, S.E., Bean, L.J., Gripp, K.W., Amemiya, A., Eds.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  6. Pierpont, E.I.; Kenney-Jung, D.L.; Shanley, R.; Zatkalik, A.L.; Whitmarsh, A.E.; Kroening, S.J.; Roberts, A.E.; Zenker, M. Neurologic and neurodevelopmental complications in cardiofaciocutaneous syndrome are associated with genotype: A multinational cohort study. Genet. Med. 2022, 24, 1556–1566. [Google Scholar] [CrossRef] [PubMed]
  7. Kenney-Jung, D.L.; Rogers, D.J.; Kroening, S.J.; Zatkalik, A.L.; Whitmarsh, A.E.; Roberts, A.E.; Zenker, M.; Gambardella, M.L.; Contaldo, I.; Leoni, C.; et al. Infantile epileptic spasms syndrome in children with cardiofaciocutanous syndrome: Clinical presentation and associations with genotype. Am. J. Med. Genet. C Semin. Med. Genet. 2022, 190, 501–509. [Google Scholar] [CrossRef]
  8. Popov, I.K.; Hiatt, S.M.; Whalen, S.; Keren, B.; Ruivenkamp, C.; van Haeringen, A.; Chen, M.-J.; Cooper, G.M.; Korf, B.R.; Chang, C. A YWHAZ Variant Associated With Cardiofaciocutaneous Syndrome Activates the RAF-ERK Pathway. Front. Physiol. 2019, 10, 388. [Google Scholar] [CrossRef]
  9. Pierpont, M.E.M.; Magoulas, P.L.; Adi, S.; Kavamura, M.I.; Neri, G.; Noonan, J.; Pierpont, E.I.; Reinker, K.; Roberts, A.E.; Shankar, S.; et al. Cardio-Facio-Cutaneous Syndrome: Clinical Features, Diagnosis, and Management Guidelines. Pediatrics 2014, 134, e1149–e1162. [Google Scholar] [CrossRef]
  10. Tartaglia, M.; Aoki, Y.; Gelb, B.D. The molecular genetics of RASopathies: An update on novel disease genes and new disorders. Am. J. Med. Genet. C Semin. Med. Genet. 2022, 190, 425–439. [Google Scholar] [CrossRef]
  11. Anastasaki, C.; Estep, A.L.; Marais, R.; Rauen, K.A.; Patton, E.E. Kinase-activating and kinase-impaired cardio-facio-cutaneous syndrome alleles have activity during zebrafish development and are sensitive to small molecule inhibitors. Hum. Mol. Genet. 2009, 18, 2543–2554. [Google Scholar] [CrossRef]
  12. Yoon, G.; Rosenberg, J.; Blaser, S.; Rauen, K.A. Neurological complications of cardio-facio-cutaneous syndrome. Dev. Med. Child Neurol. 2007, 49, 894–899. [Google Scholar] [CrossRef] [PubMed]
  13. Aizaki, K.; Sugai, K.; Saito, Y.; Nakagawa, E.; Sasaki, M.; Aoki, Y.; Matsubara, Y. Cardio-facio-cutaneous syndrome with infantile spasms and delayed myelination. Brain Dev. 2011, 33, 166–169. [Google Scholar] [CrossRef] [PubMed]
  14. Wakusawa, K.; Kobayashi, S.; Abe, Y.; Tanaka, S.; Endo, W.; Inui, T.; Iwaki, M.; Watanabe, S.; Togashi, N.; Nara, T.; et al. A girl with Cardio-facio-cutaneous syndrome complicated with status epilepticus and acute encephalopathy. Brain Dev. 2014, 36, 61–63. [Google Scholar] [CrossRef] [PubMed]
  15. Adachi, M.; Abe, Y.; Aoki, Y.; Matsubara, Y. Epilepsy in RAS/MAPK syndrome: Two cases of cardio-facio-cutaneous syndrome with epileptic encephalopathy and a literature review. Seizure 2012, 21, 55–60. [Google Scholar] [CrossRef]
  16. Battaglia, D.I.; Gambardella, M.L.; Veltri, S.; Contaldo, I.; Chillemi, G.; Veredice, C.; Quintiliani, M.; Leoni, C.; Onesimo, R.; Verdolotti, T.; et al. Epilepsy and BRAF Mutations: Phenotypes, Natural History and Genotype-Phenotype Correlations. Genes 2021, 12, 1316. [Google Scholar] [CrossRef]
  17. Iacomino, M.; Baldassari, S.; Tochigi, Y.; Kośla, K.; Buffelli, F.; Torella, A.; Severino, M.; Paladini, D.; Mandarà, L.; Riva, A.; et al. Loss of Wwox Perturbs Neuronal Migration and Impairs Early Cortical Development. Front. Neurosci. 2020, 14, 644. [Google Scholar] [CrossRef]
  18. Neuray, C.; Maroofian, R.; Scala, M.; Sultan, T.; Pai, G.S.; Mojarrad, M.; Khashab, H.E.; deHoll, L.; Yue, W.; Alsaif, H.S.; et al. Early-infantile onset epilepsy and developmental delay caused by bi-allelic GAD1 variants. Brain 2020, 143, 2388–2397. [Google Scholar] [CrossRef]
  19. Manole, A.; Efthymiou, S.; O’Connor, E.; Mendes, M.I.; Jennings, M.; Maroofian, R.; Davagnanam, I.; Mankad, K.; Lopez, M.R.; Salpietro, V.; et al. De Novo and Bi-allelic Pathogenic Variants in NARS1 Cause Neurodevelopmental Delay Due to Toxic Gain-of-Function and Partial Loss-of-Function Effects. Am. J. Hum. Genet. 2020, 107, 311–324. [Google Scholar] [CrossRef]
  20. Niestroj, L.-M.; Perez-Palma, E.; Howrigan, D.P.; Zhou, Y.; Cheng, F.; Saarentaus, E.; Nürnberg, P.; Stevelink, R.; Daly, M.J.; Palotie, A.; et al. Epilepsy subtype-specific copy number burden observed in a genome-wide study of 17 458 subjects. Brain 2020, 143, 2106–2118. [Google Scholar] [CrossRef]
  21. Alfieri, P.; Piccini, G.; Caciolo, C.; Perrino, F.; Gambardella, M.L.; Mallardi, M.; Cesarini, L.; Leoni, C.; Leone, D.; Fossati, C.; et al. Behavioral profile in RASopathies. Am. J. Med. Genet. A 2014, 164, 934–942. [Google Scholar] [CrossRef]
  22. Allanson, J.E.; Annerén, G.; Aoki, Y.; Armour, C.M.; Bondeson, M.-L.; Cave, H.; Gripp, K.W.; Kerr, B.; Nystrom, A.-M.; Sol-Church, K.; et al. Cardio-facio-cutaneous syndrome: Does genotype predict phenotype? Am. J. Med. Genet. 2011, 157, 129–135. [Google Scholar] [CrossRef] [PubMed]
  23. Rauen, K.A.; Tidyman, W.E.; Estep, A.L.; Sampath, S.; Peltier, H.M.; Bale, S.J.; Lacassie, Y. Molecular and functional analysis of a novel MEK2 mutation in cardio-facio-cutaneous syndrome: Transmission through four generations. Am. J. Med. Genet. 2010, 152, 807–814. [Google Scholar] [CrossRef] [PubMed]
  24. Steel, D.; Salpietro, V.; Phadke, R.; Pitt, M.; Gentile, G.; Massoud, A.; Batten, L.; Bashamboo, A.; Mcelreavey, K.; Saggar, A.; et al. Whole exome sequencing reveals a MLL de novo mutation associated with mild developmental delay and without “hairy elbows”: Expanding the phenotype of Wiedemann-Steiner syndrome. J. Genet. 2015, 94, 755–758. [Google Scholar] [CrossRef] [PubMed]
  25. Efthymiou, S.; Salpietro, V.; Malintan, N.; Poncelet, M.; Kriouile, Y.; Fortuna, S.; De Zorzi, R.; Payne, K.; Henderson, L.B.; Cortese, A.; et al. Biallelic mutations in neurofascin cause neurodevelopmental impairment and peripheral demyelination. Brain 2019, 142, 2948–2964. [Google Scholar] [CrossRef] [PubMed]
  26. Salpietro, V.; Efthymiou, S.; Manole, A.; Maurya, B.; Wiethoff, S.; Ashokkumar, B.; Cutrupi, M.C.; Dipasquale, V.; Manti, S.; Botia, J.A.; et al. A loss-of-function homozygous mutation in DDX59 implicates a conserved DEAD-box RNA helicase in nervous system development and function. Hum. Mutat. 2018, 39, 187–192. [Google Scholar] [CrossRef]
  27. Johnson, B.; Goldberg-Strassler, D.; Gripp, K.; Thacker, M.; Leoni, C.; Stevenson, D. Function and disability in children with Costello syndrome and Cardiofaciocutaneous syndrome. Am. J. Med. Genet. A 2015, 167, 40–44. [Google Scholar] [CrossRef] [PubMed]
  28. Papadopoulou, E.; Sifakis, S.; Sol-Church, K.; Klein-Zighelboim, E.; Stabley, D.L.; Raissaki, M.; Gripp, K.W.; Kalmanti, M. CNS imaging is a key diagnostic tool in the evaluation of patients with CFC syndrome: Two cases and literature review. Am. J. Med. Genet. A 2011, 155, 605–611. [Google Scholar] [CrossRef]
  29. Weaver, K.N.; Gripp, K.W. Central nervous system involvement in individuals with RASopathies. Am. J. Med. Genet. C Semin. Med. Genet. 2022, 190, 494–500. [Google Scholar] [CrossRef]
  30. Delogu, A.B.; Limongelli, G.; Versacci, P.; Adorisio, R.; Kaski, J.P.; Blandino, R.; Maiolo, S.; Monda, E.; Putotto, C.; De Rosa, G.; et al. The heart in RASopathies. Am. J. Med. Genet. C Semin. Med. Genet. 2022, 190, 440–451. [Google Scholar] [CrossRef]
  31. Leoni, C.; Blandino, R.; Delogu, A.B.; De Rosa, G.; Onesimo, R.; Verusio, V.; Marino, M.V.; Lanza, G.A.; Rigante, D.; Tartaglia, M.; et al. Genotype-cardiac phenotype correlations in a large single-center cohort of patients affected by RASopathies: Clinical implications and literature review. Am. J. Med. Genet. A 2022, 188, 431–445. [Google Scholar] [CrossRef]
  32. Hilal, N.; Chen, Z.; Chen, M.H.; Choudhury, S. RASopathies and cardiac manifestations. Front. Cardiovasc. Med. 2023, 10, 1176828. [Google Scholar] [CrossRef]
  33. Dentici, M.L.; Sarkozy, A.; Pantaleoni, F.; Carta, C.; Lepri, F.; Ferese, R.; Cordeddu, V.; Martinelli, S.; Briuglia, S.; Digilio, M.C.; et al. Spectrum of MEK1 and MEK2 gene mutations in cardio-facio-cutaneous syndrome and genotype–phenotype correlations. Eur. J. Hum. Genet. 2009, 17, 733–740. [Google Scholar] [CrossRef] [PubMed]
  34. Moss, C. RASopathies and the skin. Br. J. Dermatol. 2019, 180, 21. [Google Scholar] [CrossRef] [PubMed]
  35. Kavamura, M.I.; Leoni, C.; Neri, G. Dermatological manifestations, management, and care in RASopathies. Am. J. Med. Genet. C Semin. Med. Genet. 2022, 190, 452–458. [Google Scholar] [CrossRef] [PubMed]
  36. Bessis, D.; Morice-Picard, F.; Bourrat, E.; Abadie, C.; Aouinti, S.; Baumann, C.; Best, M.; Bursztejn, A.-C.; Capri, Y.; Chiaverini, C.; et al. Dermatological manifestations in cardiofaciocutaneous syndrome: A prospective multicentric study of 45 mutation-positive patients. Br. J. Dermatol. 2019, 180, 172–180. [Google Scholar] [CrossRef]
  37. Roberts, A.; Allanson, J.; Jadico, S.K.; Kavamura, M.I.; Noonan, J.; Opitz, J.M.; Young, T.; Neri, G. The cardiofaciocutaneous syndrome. J. Med. Genet. 2006, 43, 833–842. [Google Scholar] [CrossRef]
  38. Kiuru, M.; Urban, J.; Zhu, G.; Rybak, I.; Terrell, J.R.; Qi, L.; McPherson, J.D.; Marghoob, A.A.; Rauen, K.A. RAS pathway influences the number of melanocytic nevi in cardiofaciocutaneous and Costello syndromes. J. Am. Acad. Dermatol. 2020, 82, 1091–1093. [Google Scholar] [CrossRef]
  39. Leoni, C.; Guerriero, C.; Onesimo, R.; Coco, V.; Di Ruscio, C.; Acampora, A.; Esposito, I.; Romano, A.; Tartaglia, M.; Genuardi, M.; et al. Melanocytic nevi in RASopathies: Insights on dermatological diagnostic handles. J. Eur. Acad. Dermatol. Venereol. 2021, 35, e83–e85. [Google Scholar] [CrossRef]
  40. Siegel, D.H.; McKenzie, J.; Frieden, I.J.; Rauen, K.A. Dermatological findings in 61 mutation-positive individuals with cardiofaciocutaneous syndrome. Br. J. Dermatol. 2011, 164, 521–529. [Google Scholar] [CrossRef]
  41. Onesimo, R.; Sforza, E.; Giorgio, V.; Viscogliosi, G.; Kuczynska, E.M.; Margiotta, G.; Perri, L.; Limongelli, D.; Proli, F.; De Rose, C.; et al. The “FEEDS (FEeding Eating Deglutition Skills)” over Time Study in Cardiofaciocutaneous Syndrome. Genes 2023, 14, 1338. [Google Scholar] [CrossRef]
  42. Onesimo, R.; Giorgio, V.; Viscogliosi, G.; Sforza, E.; Kuczynska, E.; Margiotta, G.; Iademarco, M.; Proli, F.; Rigante, D.; Zampino, G.; et al. Management of nutritional and gastrointestinal issues in RASopathies: A narrative review. Am. J. Med. Genet. C Semin. Med. Genet. 2022, 190, 478–493. [Google Scholar] [CrossRef] [PubMed]
  43. Leoni, C.; Romeo, D.M.; Pelliccioni, M.; Di Già, M.; Onesimo, R.; Giorgio, V.; Flex, E.; Tedesco, M.; Tartaglia, M.; Rigante, D.; et al. Musculo-skeletal phenotype of Costello syndrome and cardio-facio-cutaneous syndrome: Insights on the functional assessment status. Orphanet J. Rare Dis. 2021, 16, 43. [Google Scholar] [CrossRef] [PubMed]
  44. Leoni, C.; Viscogliosi, G.; Onesimo, R.; Bisanti, C.; Massese, M.; Giorgio, V.; Corbo, F.; Tedesco, M.; Acampora, A.; Cipolla, C.; et al. Characterization of bone homeostasis in individuals affected by cardio-facio-cutaneous syndrome. Am. J. Med. Genet. A 2022, 188, 414–421. [Google Scholar] [CrossRef] [PubMed]
  45. Siano, M.A.; Pivonello, R.; Salerno, M.; Falco, M.; Mauro, C.; De Brasi, D.; Klain, A.; Sestito, S.; De Luca, A.; Pinna, V.; et al. Endocrine system involvement in patients with RASopathies: A case series. Front. Endocrinol. 2022, 13, 1030398. [Google Scholar] [CrossRef]
  46. Crincoli, E.; Leoni, C.; Viscogliosi, G.; Onesimo, R.; Mattei, R.; Tartaglia, M.; Catania, F.; Rizzo, S.; Zampino, G.; Salerni, A. Systematic ophthalmologic evaluation in cardio-facio-cutaneous syndrome: A genotype-endophenotype correlation. Am. J. Med. Genet. A 2023, 191, 2783–2792. [Google Scholar] [CrossRef]
  47. Leoni, C.; Triumbari, E.K.A.; Vollono, C.; Onesimo, R.; Podagrosi, M.; Giorgio, V.; Kuczynska, E.; Veltri, S.; Tartaglia, M.; Zampino, G. Pain in individuals with RASopathies: Prevalence and clinical characterization in a sample of 80 affected patients. Am. J. Med. Genet. A 2019, 179, 940–947. [Google Scholar] [CrossRef]
  48. Makita, Y.; Narumi, Y.; Yoshida, M.; Niihori, T.; Kure, S.; Fujieda, K.; Matsubara, Y.; Aoki, Y. Leukemia in Cardio-facio-cutaneous (CFC) syndrome: A patient with a germline mutation in BRAF proto-oncogene. J. Pediatr. Hematol. Oncol. 2007, 29, 287–290. [Google Scholar] [CrossRef]
  49. Kratz, C.P.; Franke, L.; Peters, H.; Kohlschmidt, N.; Kazmierczak, B.; Finckh, U.; Bier, A.; Eichhorn, B.; Blank, C.; Kraus, C.; et al. Cancer spectrum and frequency among children with Noonan, Costello, and cardio-facio-cutaneous syndromes. Br. J. Cancer 2015, 112, 1392–1397. [Google Scholar] [CrossRef]
  50. Niihori, T.; Aoki, Y.; Narumi, Y.; Neri, G.; Cavé, H.; Verloes, A.; Okamoto, N.; Hennekam, R.C.M.; Gillessen-Kaesbach, G.; Wieczorek, D.; et al. Germline KRAS and BRAF mutations in cardio-facio-cutaneous syndrome. Nat. Genet. 2006, 38, 294–296. [Google Scholar] [CrossRef]
  51. Al-Rahawan, M.M.; Chute, D.J.; Sol-Church, K.; Gripp, K.W.; Stabley, D.L.; McDaniel, N.L.; Wilson, W.G.; Waldron, P.E. Hepatoblastoma and heart transplantation in a patient with cardio-facio-cutaneous syndrome. Am. J. Med. Genet. A 2007, 143, 1481–1488. [Google Scholar] [CrossRef]
  52. Ney, G.; Gross, A.; Livinski, A.; Kratz, C.P.; Stewart, D.R. Cancer incidence and surveillance strategies in individuals with RASopathies. Am. J. Med. Genet. C Semin. Med. Genet. 2022, 190, 530–540. [Google Scholar] [CrossRef] [PubMed]
  53. Rauen, K.A.; Maeda, Y.; Egense, A.; Tidyman, W.E. Familial cardio-facio-cutaneous syndrome: Vertical transmission of the BRAF p. G464R pathogenic variant and review of the literature. Am. J. Med. Genet. Part A 2021, 185, 469–475. [Google Scholar] [CrossRef] [PubMed]
  54. Jurcă, M.C.; Iuhas, O.A.; Puiu, M.; Chiriţă-Emandi, A.; Andreescu, N.I.; Petcheşi, C.D.; Jurcă, A.D.; Magyar, I.; Jurcă, S.I.; Kozma, K.; et al. Cardiofaciocutaneous syndrome—A longitudinal study of a case over 33 years: Case report and review of the literature. Rom. J. Morphol. Embryol. 2022, 62, 563–568. [Google Scholar] [CrossRef] [PubMed]
  55. Wilson, B.N.; John, A.M.; Handler, M.Z.; Schwartz, R.A. Neurofibromatosis type 1: New developments in genetics and treatment. J. Am. Acad. Dermatol. 2021, 84, 1667–1676. [Google Scholar] [CrossRef] [PubMed]
  56. Sheppard, S.E.; March, M.E.; Seiler, C.; Matsuoka, L.S.; Kim, S.E.; Kao, C.; Rubin, A.I.; Battig, M.R.; Khalek, N.; Schindewolf, E.; et al. Lymphatic disorders caused by mosaic, activating KRAS variants respond to MEK inhibition. JCI Insight 2023, 8, e155888. [Google Scholar] [CrossRef]
  57. Mussa, A.; Carli, D.; Giorgio, E.; Villar, A.M.; Cardaropoli, S.; Carbonara, C.; Campagnoli, M.F.; Galletto, P.; Palumbo, M.; Olivieri, S.; et al. MEK Inhibition in a Newborn with RAF1-Associated Noonan Syndrome Ameliorates Hypertrophic Cardiomyopathy but Is Insufficient to Revert Pulmonary Vascular Disease. Genes 2021, 13, 6. [Google Scholar] [CrossRef]
  58. Andelfinger, G.; Marquis, C.; Raboisson, M.-J.; Théoret, Y.; Waldmüller, S.; Wiegand, G.; Gelb, B.D.; Zenker, M.; Delrue, M.-A.; Hofbeck, M. Hypertrophic Cardiomyopathy in Noonan Syndrome Treated by MEK-Inhibition. J. Am. Coll. Cardiol. 2019, 73, 2237–2239. [Google Scholar] [CrossRef]
  59. Wisinski, K.B.; Flamand, Y.; Wilson, M.A.; Luke, J.J.; Tawbi, H.A.; Hong, F.; Mitchell, E.P.; Zwiebel, J.A.; Chen, H.; Gray, R.J.; et al. Trametinib in Patients With NF1-, GNAQ-, or GNA11-Mutant Tumors: Results From the NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocols S1 and S2. JCO Precis. Oncol. 2023, 7, e2200421. [Google Scholar] [CrossRef]
  60. Inoue, S.; Morozumi, N.; Yoshikiyo, K.; Maeda, H.; Aoki, Y. C-type natriuretic peptide improves growth retardation in a mouse model of cardio-facio-cutaneous syndrome. Hum. Mol. Genet. 2019, 28, 74–83. [Google Scholar] [CrossRef]
  61. Inoue, S.; Moriya, M.; Watanabe, Y.; Miyagawa-Tomita, S.; Niihori, T.; Oba, D.; Ono, M.; Kure, S.; Ogura, T.; Matsubara, Y.; et al. New BRAF knockin mice provide a pathogenetic mechanism of developmental defects and a therapeutic approach in cardio-facio-cutaneous syndrome. Hum. Mol. Genet. 2014, 23, 6553–6566. [Google Scholar] [CrossRef]
  62. Rauen, K.A.; Alsaegh, A.; Ben-Shachar, S.; Berman, Y.; Blakeley, J.; Cordeiro, I.; Elgersma, Y.; Evans, D.G.; Fisher, M.J.; Frayling, I.M.; et al. First International Conference on RASopathies and Neurofibromatoses in Asia: Identification and advances of new therapeutics. Am. J. Med. Genet. A 2019, 179, 1091–1097. [Google Scholar] [CrossRef] [PubMed]
  63. Wiessner, M.; Maroofian, R.; Ni, M.-Y.; Pedroni, A.; Müller, J.S.; Stucka, R.; Beetz, C.; Efthymiou, S.; Santorelli, F.M.; Alfares, A.A.; et al. Biallelic variants in HPDL cause pure and complicated hereditary spastic paraplegia. Brain 2021, 144, 1422–1434. [Google Scholar] [CrossRef] [PubMed]
  64. Dworschak, G.C.; Punetha, J.; Kalanithy, J.C.; Mingardo, E.; Erdem, H.B.; Akdemir, Z.C.; Karaca, E.; Mitani, T.; Marafi, D.; Fatih, J.M.; et al. Biallelic and monoallelic variants in PLXNA1 are implicated in a novel neurodevelopmental disorder with variable cerebral and eye anomalies. Genet. Med. 2021, 23, 1715–1725. [Google Scholar] [CrossRef] [PubMed]
  65. Epi25 Collaborative. Sub-genic intolerance, ClinVar, and the epilepsies: A whole-exome sequencing study of 29,165 individuals. Am. J. Hum. Genet. 2021, 108, 965–982. [Google Scholar] [CrossRef]
  66. Donkervoort, S.; Kutzner, C.E.; Hu, Y.; Lornage, X.; Rendu, J.; Stojkovic, T.; Baets, J.; Neuhaus, S.B.; Tanboon, J.; Maroofian, R.; et al. Pathogenic Variants in the Myosin Chaperone UNC-45B Cause Progressive Myopathy with Eccentric Cores. Am. J. Hum. Genet. 2020, 107, 1078–1095. [Google Scholar] [CrossRef]
  67. Baldassari, S.; Musante, I.; Iacomino, M.; Zara, F.; Salpietro, V.; Scudieri, P. Brain Organoids as Model Systems for Genetic Neurodevelopmental Disorders. Front. Cell Dev. Biol. 2020, 8, 590119. [Google Scholar] [CrossRef]
  68. Dias, C.M.; Punetha, J.; Zheng, C.; Mazaheri, N.; Rad, A.; Efthymiou, S.; Petersen, A.; Dehghani, M.; Pehlivan, D.; Partlow, J.N.; et al. Homozygous Missense Variants in NTNG2, Encoding a Presynaptic Netrin-G2 Adhesion Protein, Lead to a Distinct Neurodevelopmental Disorder. Am. J. Hum. Genet. 2019, 105, 1048–1056. [Google Scholar] [CrossRef]
  69. Granata, F.; Morabito, R.; Mormina, E.; Alafaci, C.; Marino, S.; Laganà, A.; Vinci, S.L.; Briguglio, M.; Calamuneri, A.; Gaeta, M.; et al. 3T Double Inversion Recovery Magnetic Resonance Imaging: Diagnostic advantages in the evaluation of cortical development anomalies. Eur. J. Radiol. 2016, 85, 906–914. [Google Scholar] [CrossRef]
  70. Salpietro, V.; Zollo, M.; Vandrovcova, J.; Ryten, M.; Botia, J.A.; Ferrucci, V.; Manole, A.; Efthymiou, S.; Al Mutairi, F.; Bertini, E.; et al. The phenotypic and molecular spectrum of PEHO syndrome and PEHO-like disorders. Brain 2017, 140, e49. [Google Scholar] [CrossRef]
  71. Ruggieri, M.; Polizzi, A.; Schepis, C.; Morano, M.; Strano, S.; Belfiore, G.; Palmucci, S.; Foti, P.V.; Pirrone, C.; Roggini, M.; et al. Cutis tricolor: A literature review and report of five new cases. Quant. Imaging Med. Surg. 2016, 6, 525–534. [Google Scholar] [CrossRef]
  72. Pavlidou, E.; Salpietro, V.; Phadke, R.; Hargreaves, I.P.; Batten, L.; McElreavy, K.; Pitt, M.; Mankad, K.; Wilson, C.; Cutrupi, M.C.; et al. Pontocerebellar hypoplasia type 2D and optic nerve atrophy further expand the spectrum associated with selenoprotein biosynthesis deficiency. Eur. J. Paediatr. Neurol. 2016, 20, 483–488. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The RAS-MAPK pathway.
Figure 1. The RAS-MAPK pathway.
Genes 14 02111 g001
Figure 2. Neuroradiological findings in patient with CFC. Magnetic resonance imaging (MRI) scan of the brain of a 2-year-old patient with a MAP2K1 variant in CFC Syndrome. (A) Sagittal T1: Reduced cerebral volume anteriorly with associated thinning of the corpus callosum. (B) Coronal T1: Bilateral hippocampal malrotation. (C) Axial T1: Delayed myelination within the temporal lobes. (D) Axial T2: Abnormal signal within the dentate nuclei.
Figure 2. Neuroradiological findings in patient with CFC. Magnetic resonance imaging (MRI) scan of the brain of a 2-year-old patient with a MAP2K1 variant in CFC Syndrome. (A) Sagittal T1: Reduced cerebral volume anteriorly with associated thinning of the corpus callosum. (B) Coronal T1: Bilateral hippocampal malrotation. (C) Axial T1: Delayed myelination within the temporal lobes. (D) Axial T2: Abnormal signal within the dentate nuclei.
Genes 14 02111 g002
Figure 3. Distinctive craniofacial features in a CFC patient through time. At 12 months (A), 5 years (B) and 16 years of age (C,D), respectively.
Figure 3. Distinctive craniofacial features in a CFC patient through time. At 12 months (A), 5 years (B) and 16 years of age (C,D), respectively.
Genes 14 02111 g003
Table 1. Genetic findings in patients with CFC.
Table 1. Genetic findings in patients with CFC.
GeneOMIM NumberPrevalenceInheritanceCFC Phenotypic Features
BRAF*16475775%
  • De novo missense heterozygous variants
  • Autosomal-dominant missense variant inherited by mother (1 case)
Moderate to severe polymorphic seizures (+exons 11–16) 57%
Moderate ID 89%
Motor delay and hypotonia (unable to walk, needing support) 29%
Cardiac disease 72%
Greater risk of skin abnormalities
Ocular hypertelorism, optic nerve hypoplasia
Tumors (+melanoma, thyroid, colorectal, and ovarian cancers, benign nevi, premalignant colon polyps 8%
Pulmonary stenosis 50%
MAP2K1*17687225%De novo missense heterozygous variantsModerate to severe polymorphic seizures (+p.Y130C/H/N variant, exon 3) 61%
Mild ID 84%
Motor delay and hypotonia (unable to walk, needing support) 71%
Cardiac disease (lower frequency, NA)
Macrocephaly, high forehead, bitemporal constriction, hypoplasia of the supraorbital ridges, downslanting palpebral fissures
Musculoskeletal abnormalities
MAP2K2*60126325%
  • De novo missense heterozygous variants
  • Autosomal dominant missense variants inherited by mother (4 cases)
Lower risk of seizure occurrence and less severe seizure types 30%
Mild ID 25%
Motor delay and hypotonia (unable to walk, needing support) 13%
Cardiac disease 64%
Macrocephaly, high forehead, bitemporal constriction, hypoplasia of the supraorbital ridges, downslanting palpebral fissures
KRAS*1900702%
  • De novo missense heterozygous variants
  • Autosomal dominant missense variants inherited by mother (2 cases)
No epilepsy
Neurodevelopmental delay
Coarse face
Cardiac defects
YWHAZ*601288Rarely, NADe novo missense heterozygous variantsDevelopmental delay, behavioural disorders
ID
Short stature
Motor and speech delay
Triangular facies, ptosis
Seizures
Feeding problems
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Scorrano, G.; David, E.; Calì, E.; Chimenz, R.; La Bella, S.; Di Ludovico, A.; Di Rosa, G.; Gitto, E.; Mankad, K.; Nardello, R.; et al. The Cardiofaciocutaneous Syndrome: From Genetics to Prognostic–Therapeutic Implications. Genes 2023, 14, 2111. https://doi.org/10.3390/genes14122111

AMA Style

Scorrano G, David E, Calì E, Chimenz R, La Bella S, Di Ludovico A, Di Rosa G, Gitto E, Mankad K, Nardello R, et al. The Cardiofaciocutaneous Syndrome: From Genetics to Prognostic–Therapeutic Implications. Genes. 2023; 14(12):2111. https://doi.org/10.3390/genes14122111

Chicago/Turabian Style

Scorrano, Giovanna, Emanuele David, Elisa Calì, Roberto Chimenz, Saverio La Bella, Armando Di Ludovico, Gabriella Di Rosa, Eloisa Gitto, Kshitij Mankad, Rosaria Nardello, and et al. 2023. "The Cardiofaciocutaneous Syndrome: From Genetics to Prognostic–Therapeutic Implications" Genes 14, no. 12: 2111. https://doi.org/10.3390/genes14122111

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop