Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Dec 19.
Published in final edited form as: Am J Med Genet C Semin Med Genet. 2022 Dec 19;190(4):541–560. doi: 10.1002/ajmg.c.32024

New prospectives on treatment opportunities in RASopathies

Bruce D Gelb 1, Marielle E Yohe 2, Cordula Wolf 3,4, Gregor Andelfinger 5
PMCID: PMC10150944  NIHMSID: NIHMS1853821  PMID: 36533679

Abstract

The RASopathies are a group of clinically defined developmental syndromes caused by germline variants of the RAS/mitogen-activated protein (MAPK) cascade. The prototypic RASopathy is Noonan syndrome, which has phenotypic overlap with related disorders such as cardiofaciocutaneous syndrome, Costello syndrome, Noonan syndrome with multiple lentigines, and others. In this state-of-the-art review, we summarize current knowledge on unmet therapeutic needs in these diseases and novel treatment approaches informed by insights from RAS/MAPK-associated cancer therapies, in particular through inhibition of MEK1/2 and mTOR in patients with severe disease manifestations. We explore the possibilities of integrating a larger arsenal of molecules currently under development into future care plans. Lastly, we describe both medical and ethical challenges and opportunities for future clinical trials in the field.

Keywords: cardiofaciocutaneous syndrome, MEK inhibition, mTOR inhibition, Noonan syndrome, RAS-MAPK, RASopathies

1 |. INTRODUCTION

The RASopathies are a group of clinically defined developmental syndromes caused by germline mutations of the RAS/mitogen-activated protein (MAPK) cascade. The prototypic RASopathy is Noonan syndrome (NS), which is characterized by short stature, dysmorphic facial features, congenital heart disease, lymphatic disease, learning difficulties/intellectual disability, bleeding diathesis and skeletal malformations. Related disorders include cardiofaciocutaneous syndrome (CFC), Costello syndrome (CS), Legius syndrome, and NS with multiple lentigines (NSML). Typically, RASopathies are caused by activating genetic variants in the RAS/MAPK pathway, whereas NSML alleles exert their effects through an activation of the PI3K/AKT/mTOR pathway (Figure 1). RASopathies manifest with structural heart defects, specifically pulmonic stenosis, in the majority of cases. A subset of patients presents with severe hypertrophic cardiomyopathy (HCM) and lymphatic malformations, which all carry significant morbidity and mortality (Hickey et al., 2011; Wilkinson et al., 2012). Also, patients with RASopathies have a 10.5 fold higher risk than control for both childhood leukemia and solid tumors (Kratz et al., 2015).

FIGURE 1.

FIGURE 1

Patient-specific treatment concept and small molecule inhibition of RAS-MAPK pathway. Schematic depiction of the main signaling pathways in RASopathies, with gain-of-function alleles signaling culminating in increased ERK activation (right), and kinase-inactivating allele associated with NSML culminating in increased mTOR activation (left). Drugs that inhibit RAS activation include those that target RAS directly (e.g., sotorasib), inhibit RAS membrane localization (e.g., tipifarnib), prevent the activity of SHP2 (e.g., RMC-4550), inhibit the interaction of RAS with its exchange factor SOS1 (e.g., BI 1701963), or block the kinase activity of receptor tyrosine kinases (e.g., dasatinib). Please see the text for more details. Yellow font indicates preclinical tool compounds; white font indicates a drug in clinical development. Several have been FDA approved for the treatment of cancer, including sotorasib, trametinib and copanlisib, among others, and could potentially be used for the treatment of RASopathies. TTM, tetrathiomolybate. Figure created in BioRender Source: Adapted from Hebron, Hernandez, and Yohe (2022)

To date, beta-blockers remain the first line of treatment in patients with RASopathies and significant HCM (Ostman-Smith, 2014; Wolf et al., 2022). Empirical evidence points to the need of high doses (i.e., propranolol greater than 4.5 mg/kg/d) for maximum benefit in this patient population. Additional choices include L-type calcium channel blockers, disopyramide, and diuretics (Calcagni et al., 2018). For HCM in general, these medications can provide symptomatic benefits but do not reduce cardiac hypertrophy or the incidence of sudden cardiac death. For RASopathy-associated HCM, none has been rigorously tested, so efficacy for symptomatic relief is not established. Surgical options include myomectomy and, rarely, alcohol ablation (Calcagni et al., 2018; Wolf et al., 2022). Outcomes in surgical series of patients with NS are characterized by low mortality but significant morbidity, due in particular to the high risk of complete heart block in biventricular reconstruction (S. Chen et al., 2022; Hemmati et al., 2019; Laredo et al., 2018; Poterucha et al., 2015). Orthotopic heart transplantation may become necessary in RASopathy patients, but has a high mortality of 33% (McCallen et al., 2019).

Extracardiac manifestations of RASopathies also cause a significant burden of disease. Lymphovascular disease in particular is often already present prenatally and a predictor for lymphatic disease during infancy (Cox et al., 2022; Roberts, Allanson, Tartaglia, & Gelb, 2013; Sleutjes, Kleimeier, Leenders, Klein, & Draaisma, 2022; Sparks et al., 2020; Swarts et al., 2022). The lifetime prevalence of lymphatic disease is estimated at up to 37%, with age-dependent preferential manifestations: the most prevalent fetal anomalies are increased nuchal translucency and chylothorax (9% and 6%, respectively). In infants, lymphedema of the extremities (19%) and chylothorax (7%) predominate. In children and adults, lymphedema of the extremities is the most prevalent manifestation (13% and 29%, respectively; Swarts et al., 2022).

Taken together, these observations highlight the limitations of the current therapeutic arsenal for NS and related disorders and lay bare the unmet needs in the field. While the approaches described above aim at hemodynamic relief, they are not fundamentally mechanistic and fail to exploit disturbed RAS/MAPK signaling.

2 |. NEW TREATMENT OPPORTUNITIES FOR RASOPATHIES

Over the last decade, significant progress has been made in the treatment of cancers driven by gain-of-function mutations in the RAS cascade. Nineteen percent of cancer patients harbor a mutation in one of the three genes encoding RAS proteins alone, 8% harbor a mutation in BRAF, with additional mutations in other canonical RAS/MAPK components, making this cascade one of the most common targets for oncogenesis in humans (Davies et al., 2002; Prior, Hood, & Hartley, 2020). MEK1/2 occupy a crucial location in the RAS/MAPK signaling pathway and are, therefore, important targets for drug development. Over the last decade, numerous MEK inhibitors have been developed, and several of them have now been approved for oncological indications (Moore, Rosenberg, McCormick, & Malek, 2020; Zhao & Adjei, 2014). Of note, for most oncological indications, MEK inhibitors exhibit only limited efficacy as single agents and are usually combined with other anti-cancer therapeutics (Blumenschein et al., 2015; Cleary et al., 2021; Dummer et al., 2017; Kenney et al., 2021). Among various causes, this is due to the fact that inhibition of MEK1/2 can lead to the disruption of a negative feedback loop to the RAF proteins (Morrison, 2012).

Haploinsufficiency of NF1, as typically seen in pathogenic variants causing neurofibromatosis type 1, leads to a hyperactivation of the RAS-MAPK signaling cascade. In 2020, the MEK inhibitor (MEKi) selumetinib was approved as a single agent for the treatment of pediatric patients 2 years of age and older with neurofibromatosis type 1 who have symptomatic, inoperable plexiform neurofibromas (Casey et al., 2021). The overall response rate was 66%, and 82% of responding patients had a duration of response of more than 12 months. The safety profile was judged acceptable, with serious adverse events occurring in 24% of patients, mostly consisting of known MEKi class effects affecting the gastrointestinal and dermatological systems (Gross et al., 2020).

Specific treatment strategies were developed for MEKis in diverse cancer indications and have allowed the field to draw parallels and distinguish important differences with potential RASopathy-related treatment issues. Several small molecules have now been first used in adults for RAS-related cancers and then in children. The experience from this therapeutic extrapolation in hematological and oncological malignancies is encouraging to date and provides impetus to enlarge the therapeutic arsenal for disorders due to similar perturbations, such as RASopathies. In addition, the mutational spectra in RASopathies and cancer have been well studied over the last decades. Genetic studies show that the germline pathogenic variants found in RASopathies overlap minimally with the somatic mutations observed in cancer. Biochemically, this usually corresponds to a stronger activation of the cascade in cancer as compared to RASopathies, making the latter ones attractive targets for MEKi as monotherapy. Another important difference between germline RASopathies and cancer is a phenomenon called RAS addiction. This is a process in which cancers become dependent on the continued activity of oncogenes to maintain their malignant phenotype. This knowledge can be taken into account for pharmacodynamic considerations when translating therapeutic approaches from oncological applications to germline RASopathies.

3 |. INSIGHT FROM ANIMAL MODELS

Several animal models of RASopathies have been developed. Given the gain-of-function nature of RASopathy alleles, knock-in mice carrying specific disease-causing variants are of particular relevance for this review. Several mouse models carrying such genetic variants have been engineered that recapitulate aspects of RASopathy phenotypes, including short stature, altered craniofacial development and hematologic anomalies. While no mouse model fully mimics the human phenotype of obstructive HCM, a number of these models manifest at least some degree of cardiac hypertrophy (RAF1 L613V, RIT1 M90I, RIT1 A57G, KRAS V14I, HRAS G12S, KRAS T58I; Castel et al., 2019; Hernandez-Porras et al., 2014; Oba et al., 2018; Takahara et al., 2019; Wong et al., 2020; Wu et al., 2011). In two of them, modulation of the RAS/MAPK cascade using the MEK inhibitor PD0325901 ameliorated the cardiac phenotype: in the RAF1 L613V mouse, treatment was started at 1 month of age and pursued for 6 weeks, resulting in prevention or reversal of cardiac hypertrophy (Wu et al., 2011). In the KRAS V14I mouse, responses varied according to age at which treatment was started. Beneficial effects on cardiac hypertrophy and survival were more pronounced with earlier treatment, and persisted after cessation of treatment (Hernandez-Porras et al., 2014). In a mouse model of NSML with the PTPN11 Y279C allele, treatment with the AKT inhibitor ARQ 092 at 12 weeks of age reduced cardiac hypertrophy and normalized heart size after 4 weeks of treatment (J. Wang et al., 2017). Similar results with reversal of cardiac hypertrophy were obtained in this model using rapamycin to inhibit mTOR (Marin et al., 2011). Low-dose dasatinib, a Bcl-Abl and Src kinase family inhibitor, ameliorates HCM in this same model (Yi et al., 2022). In aggregate, the findings summarized above provide the rationale for implementing patient-specific, mechanistic therapies to improve the most critical manifestations of RASopathies.

4 |. MEETING AN UNMET NEED: FIRST EXPERIENCES FROM COMPASSIONATE USE OF MECHANISTIC THERAPIES

The first extrapolation of mouse studies to human pathology occurred with the administration of the mTOR inhibitor (mTORi) everolimus in a young infant with NSML and severe HCM (Figure 1; Hahn et al., 2015). Exploiting the experimental evidence that these missense substitutions activate the PI3K/AKT/mTOR pathway rather than the RAS/MAPK/ERK cascade, a critically ill neonate with a PTPN11 Q510E allele and HCM received everolimus from the age of 24 weeks onwards (Hahn et al., 2015). In this case, significant clinical improvement was seen while awaiting heart transplantation, which occurred at 36 weeks (i.e., after 12 weeks of treatment). However, there was still severe HCM at the time of surgery, and histologic examination of the myocardium showed the typical findings observed in HCM. Whether the everolimus treatment resulted in the reduced heart failure and whether or not the HCM might have diminished with a longer treatment course is unclear.

When confronted with two infants with RIT1 pathogenic variants and severe HCM with heart failure, we (G.A. and B.D.G.) postulated a class effect for MEKis in RASopathies (i.e., an FDA-approved MEKi would be as effective as PD0325901) and that regression of HCM would be possible with such treatment in humans (Figure 1; Andelfinger et al., 2019). For both patients, conventional therapies had been exhausted, and disease progression made an unfavorable outcome highly likely. Therefore, both infants were started at the age of 3 months on the MEKi trametinib, which was FDA approved for melanoma. Due to the lack of data about pharmacokinetics and side-effect profile in infants for this drug, strict surveillance for potential side effects was implemented, including monitoring for retinal damage (which has been reported in adults with cancer treated with trametinib). Second, we consulted oncologists, pediatric oncologist, pharmacologist and pharmacists with expertise in small molecular kinase inhibitors and drug monitoring and related all known information to the parents. Also, appropriate clinical ethics approval and informed consent from the parents was also obtained. This consent also considered the possibility that novel, unknown effects could occur during the treatment. Third, a pre-defined time threshold of 3 months was set by which point clinical improvement had to be present or treatment would be discontinued. Fourth, exploiting the insight from biochemical study of RASopathy mutations, a dosing regimen below oncological dosing was chosen empirically. This key postulate, which takes into account cellular state rather than disease severity, is depicted in Figure 2. In essence, we predict that the higher biochemical signaling strength of cancer-associated RAS mutations will require higher doses of MEK inhibition; conversely, for RASopathy patients, lower doses should suffice. Whether this quantitative concept developed from biochemical studies will also hold true in practice needs to be determined in future clinical trials.

FIGURE 2.

FIGURE 2

Rationale for low-dose MEK inhibition in RASopathies. Left column: normal signaling strength in a healthy individual as depicted by an intermediate gray level in all cells of the body (top and middle row). Middle column: Homogenously increased ERK signaling strength in a germline RASopathy (top row), uniformly decreased signaling strength after mild MEK inhibition (middle row). For the purpose of illustrating an effect on hypertrophic cardiomyopathy, the heart is highlighted as a target organ. Right column: Very strongly increased ERK signaling strength in a somatic mutation of a tumor, with normal ERK signaling strength in the unaffected cells of the body (top row), inhomogenously decreased ERK signaling after strong MEK inhibition (middle row). Bottom row: Gradient indicating degree of ERK signaling strength. For more detailed discussion of mechanisms and caveats see text

In both children, rapid and sustained improvement of cardiac status was observed in the following sequences: clinical status within the first week, neurohumoral improvement with decrease of amino-terminal-pro-brain natriuretic peptide (NT-pro-BNP) within 4–6 weeks, and echocardiographic improvement at 6–12 weeks (Figures 3 and 4). In both patients, subvalvular and valvular obstruction of the outflow tracts occurred (Figure 3). Of note, in one patient, resolution of ventricular arrhythmia and lymphatic drainage was also noted (Andelfinger et al., 2019). Based upon the observation that most of the attrition in RASopathy with severe HCM occurs during the first 2 years of life and adding a security window, we initially limited duration of treatment for these first two patients to 2½ years. Signs of a relapse were present 1 month after treatment cessation, with increases in NT-pro-BNP and wall thicknesses in both patients, so they were treated for one more year with MEKi. A second attempt to wean at this time was successful, and after more almost 2 years off therapy, both patients continue to do well, with normal cardiac status and without known side effects of MEKi (Figures 3 and 4).

FIGURE 3.

FIGURE 3

Effect of MEKi on (sub)valvular obstruction. Response to treatment with trametinib in a patient with biventricular outflow tract obstruction due to a RIT1 S35T mutation. Evolution from birth (first row, mild valvular pulmonary stenosis, gradient 22 mmHg), start of treatment at 3 months of age (second row, severe, mainly infundibular obstruction, 82 mmHg), after 9 months of treatment (third row, mild valvular pulmonary stenosis, 30 mmHg), after 2 years of treatment (bottom row, no gradient)

FIGURE 4.

FIGURE 4

Effect of MEKi on HCM. All pictures are end-diastolic frames in short axis view from the same patient as in Figure 3. (a) Marked concentric hypertropy at 3 months of age, (b and c) improvement after 3 months and 1 year of treatment, respectively, (d) normal ventricular morphology 1 year after treatment cessation

Subsequent to the publication of the mechanistic treatment of two infants with NS-related HCM with trametinib, several other publications described experiences with MEKi for severe manifestations in RASopathies (Dori et al., 2020; Li et al., 2019; Meisner, Bradley, & Russell, 2021; Mussa et al., 2021; Nakano et al., 2022). Excluding our initial study, the published evidence now reports amelioration in three additional cases with cardiovascular issues (Lioncino et al., 2022; Meisner et al., 2021; Mussa et al., 2021). In two of these cases, the leading symptom was intractable multifocal atrial arrhythmia, which improved within 48 hours of introduction of a MEKi without appreciable conduction anomalies (Lioncino et al., 2022; Meisner et al., 2021). In the third case, which described a multimorbid premature newborn, the cardiopulmonary status improved to a point that allowed extubation, however, acute decompensation after a surgical procedure for hydrocephalus resulted in an intractable relapse of HCM despite continuation of the MEKi. In this patient, concomitant pulmonary vascular disease was suspected clinically and confirmed at autopsy. It is, therefore, conceivable that MEK inhibition cannot reverse pulmonary vascular disease to the same degree or in all patients with NS and pulmonary involvement (Mussa et al., 2021). A total of six patients with NS and severe lymphovascular disease have now been reported as well in addition to a report of two patients with ARAF pathogenic variants and central conducting lymphatic anomaly (this latter condition is extremely rare and has not usually been categorized as a RASopathy) (Dori et al., 2020; D. Li et al., 2019; Lioncino et al., 2022; Nakano et al., 2022). In all of these patients, significant improvement or resolution of lymphovascular disease was reported, with initial response starting at around 2 days to 1 month after treatment initiation to sustained response at 12 months after treatment initiation (example in Figure 5).

FIGURE 5.

FIGURE 5

Effect of MEKi on lymphovascular anomalies. Magnetic resonance lymphangiography (a) before and (b) after treatment with trametinib showing near complete resolution of left-sided lung perfusion (arrow) and disappearance of large left-sided ducts Source: Reproduced with permission from Dori et al. (2020)

We note that these reports now cover both pathways known to be involved in RASopathy, across a wide variety of genotypes, and specific time windows for response (Figure 6). We, therefore, conclude that these results allow extrapolation from experience in oncology and animal models to treatment strategies in RASopathies, despite some differences of phenotypic manifestation of RASopathies in genetically engineered mouse models. These differences are as follows: (a) while the HCM observed in mouse models can be eccentric, mouse models of RASopathies do not show obstruction of the outflow as found in humans and (b) while prenatal edema is frequently noted in RASopathy mouse models, there are no adequate mouse models of postnatal lymphovascular disease.

FIGURE 6.

FIGURE 6

Temporal pattern of response to MEKi. Blue gradient arrows depict gradual onset and duration of response to MEKi for different organ systems. Adverse effects can occur at any time. Based upon cited literature and unpublished data

In summary, it is noteworthy that the majority of the reports of targeted inhibition of the RASopathy-related signaling events with MEKis or mTORis in severely or critically ill patients with cardiac or lymphovascular involvement is positive. For some patients, this is true even in situations where one can reasonably anticipate that conventional therapies would have failed. Also, no death attributable to medication has been reported, even for very sick children. Overall, toxicity seems to be reasonably well tolerated and manageable in most cases. Efforts to collect larger series of these infants and children are currently ongoing in the field.

5 |. ONE SIZE WILL NOT FIT ALL: WIDENING OPTIONS TO MODULATE RAS/MAPK SIGNALING

As noted above, many novel agents designed for the treatment of cancers driven by alterations in the RAS/MAPK pathway have been developed recently. These agents could represent treatment options for patients with RASopathies with the exception of NSML (Hebron et al., 2022). The novel agents fall into three classes, outlined in more detail below: inhibitors of RAS activation, direct RAS inhibitors, and inhibitors of the MAPK pathway (Moore et al., 2020). In large part, these agents have not yet been tested in animal models of RASopathies but have the potential to be more efficacious or cause fewer side effects than MEK inhibitors in these models (Figure 1).

5.1 |. Inhibitors of RAS activation

Several classes of drugs can be categorized as inhibitors of RAS activation (Table 1). These include prenylation inhibitors, SHP2 inhibitors, and SOS1 inhibitors. C-terminal prenylation is required for RAS membrane localization and activation. Both the HMG CoA-reductase inhibitors (e.g., atorvastatin, lovastatin) and the N-bisphosphonates (e.g., alendronate, pamidronate) inhibit the synthesis of substrates used in the prenylation reaction. Either HMG CoA-reductase inhibitors or N-bisphosphonates, then, might prevent RAS prenylation and, thus, activation in patients with RASopathies (Baranyi, Buday, & Hegedus, 2020). These drugs are well studied and relatively well tolerated when used to treat hypercholesterolemia and osteoporosis in adults and children, and, thus, represent potential long-term treatment options for patients with RASopathies. Importantly, lovastatin reverses learning defects in a mouse model of NS, Ptpn11D61G/+ (Lee et al., 2014). Based on these results, a phase 3 clinical trial of simvastatin for the treatment of growth and bone abnormalities in children with NS is currently underway (NCT02713945). Neither the HMG CoA-reductase inhibitors nor the N-bisphosphonates have been evaluated in other preclinical RASopathy models.

TABLE 1.

Compounds of interest in RASopathies

Drug Mechanism of action Indications Typical adverse events Comments References
Dasatinib Tyrosine kinase inhibitor (BCR-Abl, Src, c-Kit, ephrin receptors, others) Philadelphia-chromosome positive CML and ALL (a)
Noonan syndrome-associated hypertrophic cardiomyopathy (o)
Fluid retention/edema, hemorrhage, musculoskeletal pain, myelosuppression, pleural effusion Tested in PTPN11 D61G and PTPN11 Y279C Noonan syndrome mouse models (Yi et al., 2016; Yi et al., 2022)
BI 1701963 SOS1::panKRAS inhibitors Solid tumors with KRAS mutation (t) n/a Binds to the catalytic domain of SOS1, conferring pan-KRAS inhibition (Parikh et al., 2022)
Adagrasib KRAS G12C inhibitor, allele-specific, locks KRAS G12C in inactive state KRAS G12C-mutated cancers (t) Diarrhea, nausea, vomiting, fatigue, alanine transaminase increase, blood creatinine increase, aspartate aminotransferase increase, decreased appetite KRAS G12C not found as germline mutation (Kessler et al., 2020; Nagasaka et al., 2020; Parikh et al., 2022)
ASO/SSO Anti-sense oligonucleotides and splice-switching oligonucleotides In principle, all KRAS mutant alleles (t) n/a To date, 13 NDAs granted by FDA for oligonucleotide therapies, none in cancer (Crooke, Baker, Crooke, & Liang, 2021; Kamerkar et al., 2017; Ross et al., 2017; D. Wang et al., 2022)
BI 2852 KRAS G12D inhibitor KRAS G12D-mutated cancers (t) n/a KRAS G12D not found as germline mutation. (Kessler et al., 2019)
Sotorasib KRAS G12C inhibitor, allele-specific, locks KRAS G12C in inactive state KRAS G12C-mutated cancers (a) Diarrhea, musculoskeletal pain, nausea, fatigue, hepatotoxicity, cough, decreased lymphocytes, decreased hemoglobin, increased aspartate aminotransferase, increased alanine aminotransferase, decreased calcium, increased alkaline phosphatase First FDA-approved oral KRAS G12C inhibitor
KRAS G12C not found as germline mutation
(Kessler et al., 2020; Nagasaka et al., 2020; Parikh et al., 2022)
Alpelisib α-Specific PI3K inhibitor Metastatic breast cancer (a), PIK3CA-related overgrowth spectrum (PROS) who require systemic therapy (a) Diarrhea, stomatitis, hyperglycemia, cutaneous adverse reactions, pneumonitis Inhibits p110α approximately 50 times more than other PI3K isoforms (Fritsch et al., 2014; Ross et al., 2017; Venot et al., 2018)
Copanlisib α- and δ-specific PI3K inhibitor Relapsed follicular lymphoma (a), marginal zone lymphoma (o) Serum enzyme elevations, liver failure Inhibits all four class I isoforms, tenfold higher for p110α and p110δ (Krause, Hassenruck, & Hallek, 2018)
Idelalisib Preferential PI3Kδ inhibitor Relapsed/refractory chronic lymphocytic leukemia (CLL, in combination with rituximab) (a), relapsed follicular B-cell non-Hodgkin lymphoma (a), relapsed small lymphocytic leukemia (a) Hepatic toxicities, severe diarrhea, colitis, pneumonitis, infections, and intestinal perforation Affinity to PI3Kδ>PI3Kβ>PI3Kα in a ratio of 4:2:1 (Lannutti et al., 2011; Yang, Modi, Newcomb, Queva, & Gandhi, 2015)
Samotolisib Triple PI3K, mTOR, and DNA-dependent protein kinase, catalytic subunit (PRKDC) inhibitor Relapsed or refractory advanced solid tumors, non-Hodgkin lymphoma, or histiocytic disorders with TSC or PI3K/MTOR mutations (all t) Leukopenia, neutropenia, thrombocytopenia, nausea, fatigue, diarrhea Inhibits all PI3K isoforms (α, β, δ, γ) (Smith et al., 2016)
Capivasertib Pan-AKT inhibitor Relapsed or refractory B-cell non-Hodgkin lymphoma (t), triple negative breast cancer, HR+/HER2− breast cancer, metastatic castration resistant prostate cancer, prostate cancer with PTEN loss, endometrial cancer, meningioma, cancers with AKT changes, recurrent ovarian cancer (all t) Diarrhea, nausea, hyperglycemia, vomiting, rash Binds to and inhibits all AKT isoforms (B. R. Davies et al., 2012; Uko, Guner, Matesic, & Bowen, 2020)
Ipatasertib Pan-activated AKT inhibitor Breast cancer, solid tumors, solid tumors with AKT mutations, non-small cell lung cancer, triple-negative breast cancer, endometrial cancer, fallopian tube carcinoma, ovarian carcinoma, prostate cancer, glioblastoma, gastric cancer, gastroesophageal junction cancer, cancer of unknown primary site (all t) Diarrhea, fatigue, vomiting, neutropenia Exhibits differential activity in cells with and without activated Akt signaling, distinct from phosphoinositide 3-kinase (PI3K) inhibitors (Lin et al., 2013; Uko et al., 2020)
Miransertib Pan-AKT inhibitor PROS (c) and (t) Dry mouth, mucositis, pharyngitis, sinus tachycardia, headache, pain Currently no active trials in oncology
Tested in PTPN11 Y279C mouse model
(Forde et al., 2021; Kobialka et al., 2022; Leoni et al., 2019; Ours, Sapp, Hodges, de Moya, & Biesecker, 2021; Yu et al., 2015)
MK-2206 Pan-AKT inhibitor  Non-small cell lung cancer, solid tumors, ovarian sarcoma, recurrent Fallopian tube and ovarian carcinoma, recurrent primary peritoneal carcinoma, oral and salivary gland carcinoma, squamous cell carcinoma of the nasopharynx, colorectal carcinoma, endometrial carcinoma, breast cancer, lymphoma, gastric carcinoma, pancreatic and neuroendocrine tumors, renal cell carcinoma, prostate cancer (all t)  Diarrhea, rash, nausea, fatigue, hyperglycemia  Inhibits AKT membrane localization (Uko et al., 2020)
Everolimus mTOR inhibitor Immunosuppression (a), tuberous sclerosis complex (TSC) associated seizures (a) and tumors (a,c), advanced renal cell carcinoma (a), subependymal giant cell astrocytoma associated with TSC (a) Stomatitis, infections, asthenia, fatigue, cough, diarrhea, upper respiratory tract infection, sinusitis, otitis media, fever Allosteric mTOR inhibitor
Selective for mTORC1 without activation of mTORC2
Compassionate use in one case of NSML with HCM described
(Y. Chen & Zhou, 2020; Hahn et al., 2015; Witzig et al., 2015)
Sirolimus mTOR inhibitor Immunosuppression (a), lymphangioleiomyomatosis (a), bone sarcoma (o), tuberous sclerosis complex (TSC) (o), chronic non-infectious uveitis (o), pachyonychia congenital (o), angiofibromas (o), beta-thalassemia (o), pulmonary arterial hypertension (o), sickle cell disease (o), familial adenomatous polyposis (o) Peripheral edema, hypertriglyceridemia, hypertension, hypercholesterolemia, increased creatinine, stomatitis, abdominal pain, nausea, diarrhea, headache, dizziness, fever, urinary tract infection, anemia, arthralgia, pain, thrombocytopenia, nasopharyngitis, acne, chest pain, upper respiratory tract infection, myalgia Allosteric mTOR inhibitor
Compassionate use in one case of lymphatic malformation with KRAS G12D mutation
(Y. Chen & Zhou, 2020; Sideris, Tng, & Chee, 2021)
Temsirolimus mTOR inhibitor Advanced renal cell carcinoma (a), lymphoma, approximately 1,000 clinical studies with a variety of malignancies Hypersensitivity reactions, oral ulcers, diarrhea, nausea, poor appetite, fatigue, peripheral edema, rash, anemia Intravenous application only (Y. Chen & Zhou, 2020)
JAB-3068 SHP2 inhibitor Non-small cell lung cancer, head and neck cancer, esophageal cancer, advanced solid tumors, other metastatic solid tumors (t), esophageal cancer (o) No data
RLY-1971 Allosteric SHP2 inhibitor Advanced or solid metastatic tumors (t) No data Binds and stabilizes SHP2 in its inactive conformation (Williams et al., 2022)
RMC-4550 Allosteric SHP2 inhibitor Shows in vitro efficacy against lung adenocarcinoma and BRAF-, NF1- and RAS-driven cancer cell lines No data (Adamopoulos et al., 2021; Nichols et al., 2018; R. R. Wang, Liu, Zhou, Ma, & Wang, 2020; Zhou et al., 2022)
TNO155 Allosteric SHP2 inhibitor EGFR- and KRAS G12C mutant non-small cell lung cancer, squamous cell carcinoma, gastrointestinal stromal tumors, colorectal cancer, BRAF V600 positive colorectal cancer Increased creatine kinase, peripheral edema, diarrhea, acneiform dermatitis (LaMarche et al., 2020)
Alendronate Inhibits farnesyl diphosphate synthase Currently no ongoing clinical studies in RAS/MAPK related pathologies Abdominal pain, acid regurgitation, constipation, diarrhea, dyspepsia, musculoskeletal pain, nausea Inhibits RAS farnesylation and thus membrane targeting of RAS (Luckman et al., 1998)
Lovastatin HMG-CoA reductase inhibitor Growth and bone abnormalities in Noonan syndrome (t) Blurred vision, constipation, dyspepsia, abdominal pain, diarrhea, flatulence, nausea, myalgia, muscle spasms, dizziness, headache, skin rash Inhibits RAS farnesylation and thus membrane targeting of RAS
Lovastatin ameliorates cognitive defects in Nf1 and Ptpn11 mice, but not HRAS mice. Findings not replicated in children with NF1
(Krab et al., 2008; Lee et al., 2014; W. Li et al., 2005; Payne et al., 2016; Schreiber et al., 2017)
Tipifarnib Farnesyltransferase inhibitor NF1-associated progressive plexiform neurofibromas (t), HRAS-mutated head and neck squamous cell carcinoma (b), solid tumors with HRAS gene alterations, leukemia, relapsed/refractory T cell lymphoma, relapsed/ refractory myeloma, glioblastoma, astrocytoma, oligodendroglioma, recurrent breast cancer, metastatic malignant melanoma, advanced pancreatic cancer, bladder cancer (all t) CK elevation; gastrointestinal toxicities (abdominal pain, diarrhea, constipation, nausea, vomiting, anorexia, oral mucositis, sore throat, and dry mouth), acneiform rash, maculopapular rash Inhibits RAS farnesylation and thus membrane targeting of RAS
Possible HRAS preference, since HRAS is uniquely dependent on farnesyl transferase for prenylation (NRAS and KRAS can bypass via geranylgeranyl transferase)
(End et al., 2001; Kessler et al., 2019; Lee et al., 2014; Sebti & Hamilton, 2000; Sepp-Lorenzino et al., 1995)
Tovorafenib Type II pan-RAF inhibitor Pediatric low-grade glioma (b), malignant glioma (o), Langerhans cell histiocytosis, low-grade glioma, craniopharyngioma (all t) Increased serum creatine kinase, rash, hair color change High brain penetrance. Little paradoxical MEK activation. (Sun et al., 2017)
Belvarafenib Type II pan-RAF inhibitor NRAS-mutated advanced melanoma, solid tumors (all t) Rash, acneiform dermatitis, pyrexia Stabilizes RAF in its inactive conformation and inhibits RAF dimerization. Little paradoxical MEK activation (Adamopoulos et al., 2021; Moore et al., 2020; Yen et al., 2021)
LY3009120 Type II pan-RAF inhibitor Solid tumors, melanoma, non-small cell lung cancer, colorectal cancer (all t) Fatigue, nausea, acneiform dermatitis, maculopapular rash, arthralgia, myalgia Inhibits all RAF isoforms, inhibits BRAF and CRAF homodimers and heterodimers. Little paradoxical MEK activation (Adamopoulos et al., 2021; Lai et al., 2022; Moore et al., 2020; Peng et al., 2015; Sullivan et al., 2020)
LXH254 Selective RAF inhibitor Solid tumors, non-small cell lung cancer, ovarian cancer, melanoma, colorectal cancer Acneiform dermatitis, maculopapular rash, fatigue, nausea, myalgia, increased lipase Potent inhibitor of BRAF and CRAF, less activity against ARAF (Janku et al., 2018; Monaco et al., 2021; Ramurthy et al., 2020)
Binimetinib MEK inhibitor BRAF V600E or V600K mutation-positive unresectable or metastatic melanoma (a)
Non-small cell lung cancer with KRAS gene mutation, craniopharyngioma, BRAF mutant cancer, hairy cell leukemia, gastrointestinal stromal tumor, pancreatic cancer, colorectal cancer, biliary tract cancers, melanoma, pancreatic carcinoma, solid tumors with RAS alterations, breast cancer, ovarian cancer, fallopian tube cancer (all t)
Fatigue, nausea, diarrhea, constipation, vomiting, abdominal pain, acneiform dermatitis, retinopathy Used in combination with encorafenib (Dummer et al., 2018)
Cobimetinib MEK inhibitor BRAF V600E or V600K mutation-positive unresectable or metastatic melanoma (a)
Extracranial arteriovenous malformations (t)
Histiocytosis, non-small cell lung cancer, solid tumors, recurrent acute myeloid leukemia, squamous cell carcinoma, adenocarcinoma, pancreatic cancer, ovarian cancer, biliary tract carcinoma, thyroid carcinoma (all t)
Diarrhea, photosensitivity, nausea, fever, vomiting, acneiform dermatitis, impaired vision, retinopathy, retinal detachment Used in combination with vemurafenib
Positive responses reported in plexiform neurofibroma in neurofibromatosis type 1 and a case of dysembryoplastic neuroepithelial tumor in Noonan syndrome
(Larkin et al., 2014; Trippett et al., 2022)
Mirdametinib MEK inhibitor NF1-associated plexiform neurofibromas (f)
Breast cancer, low grade glioma, advanced or refractory solid tumors (all t)
Rash, fatigue, nausea, vomiting Oncological use planned with RAF dimer inhibitor (de Blank et al., 2022; Klesse et al., 2020; Weiss et al., 2021)
Selumetinib MEK inhibitor NF1-associated plexiform neurofibromas (a)
Melanoma, Kaposi’s Sarcoma, blood cancers, solid tumors, breast cancer, ovarian cancer, endometrial cancer, non-squamous cell lung cancer, non-small cell lung cancer, sarcoma, pancreatic cancer, biliary tract cancer, colorectal cancer, gastric adenocarcinoma, pancreatic carcinoma, hepatocellular carcinoma, bladder cancer, vestibular schwannoma, thyroid cancer
Vomiting, rash, abdominal pain, diarrhea, nausea, dry skin, fatigue, musculoskeletal pain, stomatitis, blurred vision, retinal pigment and epithelial detachment, paronychia One compassionate use case in Noonan syndrome associated bleeding described (Casey et al., 2021; Chakraborty et al., 2022; de Blank et al., 2022; Gross et al., 2020; Kenney et al., 2021)
Trametinib MEK inhibitor BRAF V600E or V600K mutation-positive unresectable or metastatic melanoma (a), metatstatic non-small lung cancer with BRAF V600E mutation (a), anaplastic thyroid cancer with V600E mutation (a), metastatic or unresectable tumors with BRAFV600E mutation (a)
Extracardiac arterio-venous malformation (t)
Non-small cell lung cancer, melanoma, solid tumors, pancreatic cancer, squamous cell cancer, cervical cancer, thyroid cancer, juvenile myelomonocytic leukemia, acute myeloid leukemia, lymphoma, hemangioendothelioma, meningioma, breast cancer, colorectal cancer, prostate cancer, renal cancer, craniopharyngioma, astrocytoma, Erdheim-Chester disease (all t)
Rosacea (topical)
Diarrhea, constipation, acneiform rash, serous retinopathy, retinal vein occlusion, uveitis, interstitial lung disease, hemorrhage, venous thromboembolism, hypertension Oncological use in combination with dabrafenib
Positive responses in germline RASopathy-associated pathologies (see main article)
(de Blank et al., 2022; Kondyli et al., 2018; Moore et al., 2020; Robert et al., 2015; Roskoski, 2018)
Tetrathiomolybdate (TTM) Copper chelator Wilson’s disease (a in Europe)
Non-small cell lung cancer, prostate cancer, breast cancer, esophageal carcinoma, biliary tract cancer, breast cancer, colorectal cancer, multiple myeloma (t)
Anemia, leukopenia, thrombocytopenia Restricts copper binding of MEK1/2, which is required for MEK1-dependent phosphorylation of ERK (Brady et al., 2014; Turski et al., 2012; Xu, Casio, Range, Sosa, & Counter, 2018)
Ulixertinib ERK inhibitor BRAF mutant melanoma (o) (Germann et al., 2017)

Note: Summary of selected compounds that are in clinical development in oncology or RAS-MAPK driven diseases which could be of potential interest for future application in germline RASopathies. Indications and adverse effects are not exhaustive. Abbreviations: (a) approved indication(s)/mutation(s), (b) breakthrough therapy designation, (c) compassionate use, (f) fast track designation. (o) orphan drug designation, (t) targeted indication(s)/mutation(s).

The farnesyl transferase inhibitors (FTIs, such as tipifarnib and lonafarnib), directly inhibit the enzyme responsible for a particular type of RAS prenylation, namely farnesylation. NRAS and KRAS bypass the requirement for farnesylation by undergoing geranyl geranylation, an alternative prenylation that allows for membrane localization. However, HRAS, the RAS isoform affected in CS, is unable to be alternatively modified, so its membrane localization and cellular function are suppressed by FTIs (Cox, Der, & Philips, 2015). FTIs, then, may represent a potential therapeutic for CS, but not the KRAS- or NRAS-altered RASopathies.

Two classes of targeted agents that prevent RAS activation recently have been identified: SOS1 inhibitors and SHP-2 inhibitors (SHP-2 is encoded by PTPN11). SOS1 inhibitors (e.g., BI 1701963 and RMC-5845) that inhibit the interaction between RAS proteins and its exchange factor SOS1 to inhibit RAS activation are in clinical development for RAS-mutated malignancies (Hillig et al., 2019). SOS inhibitors have not yet been evaluated in models of RASopathies but could be expected to have efficacy for any genotype that requires the RAS activation cycle to be intact to achieve aberrant signaling through the MAPK pathway. Allosteric inhibitors of SHP-2, which hold the phosphatase in the auto-inhibited conformation, are also in clinical trials in malignancies (Yuan, Bu, Zhou, Yang, & Zhang, 2020). These inhibitors, including RMC-4630, TNO155, and RLY-1971, are unable to bind to SHP-2 mutants in which the auto-inhibited conformation is destabilized (Padua et al., 2018); therefore, these inhibitors are unlikely to be active for most PTPN11 alleles in NS, which do perturb SHP-2 auto-inhibition. This limitation may be prevented in a novel approach that targets the N-terminal Src homology domain of SHP2, allowing mutant SHP2 to be targeted (Bobone et al., 2021). Through their inhibitory role on the nexus of RAS exchange, RMC-4630 and related SHP-2 inhibitors may be effective in RASopathies caused by variants in genes other than PTPN11, although this hypothesis has not yet been tested.

5.2 |. Direct RAS inhibitors

Direct inhibitors of KRASG12C (e.g., AMG 510, MRTX849, JNJ-74699157, and GDC-6036) function by binding covalently to KRASG12C-GDP, locking it in the inactive state. These inhibitors are currently in clinical development for KRASG12C-mutated solid tumors (Moore et al., 2020), and AMG 510 has been FDA approved for the treatment of KRASG12C-mutated non-small cell lung carcinoma based on results from the CodeBreak 100/101 trial (Hong et al., 2020). These inhibitors are an exciting advance in the RAS field, as it was previously thought that direct RAS inhibition was impossible. However, the KRASG12C inhibitors are not likely to have utility in individuals with RASopathies because the KRASG12C variant is uncommon in the RASopathies. Drugs that specifically inhibit other KRAS mutants, such as KRASG12D (MRTX1133, KRpep-2D) (Lim et al., 2021; Wang et al., 2022) and KRASG12S (Zhang, Guiley, & Shokat, 2022) are being developed and may have clinical utility in patients with KRAS-mutant NS or CFC in the future.

Strategies to target KRAS in a mutation-independent manner are also being developed. BI 2825, for example, binds to a grove on the surface of both active and inactive KRAS (Kessler et al., 2019). Additional preclinical tool compounds, RMC-6236 and RMC-6291, are pan-RAS inhibitors that bind to a complex of RAS-GTP (RAS ON) and the ubiquitously expressed molecular chaperone cyclophilin A (Koltun et al., 2022; Nichols et al., 2022). Agents that modify the expression of RAS, such as antisense (Ross et al., 2017) and splice switching (Hartung et al., 2016) oligonucleotides, have also been developed. Clinical candidates based on any of these mutation-independent agents represent exciting potential therapeutics for RASopathies of several genotypes.

5.3 |. RAS/MAPK pathway inhibitors

As discussed above, MEKis are in clinical evaluation for the treatment of RASopathy-related hypertrophic cardiomyopathy. Agents in addition to MEKis also inhibit signaling through the RAS/MAPK pathway. For example, the MEK1/2 kinases require the heavy metal copper for their kinase activity. Copper chelators, which are well tolerated clinically, are efficacious in preclinical models of RAS/MAPK-driven malignancies and might represent a novel method by which to decrease MAPK signaling in individuals with RASopathies (Baldari, Di Rocco, & Toietta, 2020). Importantly, the tool compound U0126, which functions as a MEKi in part due to its ability to chelate copper, is effective in several RASopathy models (Jaffre et al., 2019; Josowitz et al., 2016; Nakamura, Gulick, Pratt, & Robbins, 2009; Rodriguez-Viciana et al., 2006; Senawong et al., 2008).

Pan-RAF inhibitors that are in clinical development, such as LY3009120, LXH254, DAY101 (also known as MLN2480 or TAK-580), and belvarafenib, are able to inhibit MAPK signaling in the absence of BRAFV600E, while BRAFV600E-specific inhibitors, such as vemurafenib and dabrafenib, cause paradoxical activation of the MAPK pathway in the absence of BRAFV600E (Durrant & Morrison, 2018). BRAFV600E alleles have not been associated with RASopathies to date; therefore, the pan-RAF inhibitors may have more clinical utility than BRAFV600E inhibitors in patients with RASopathies. ERK inhibitors, such as ulixertinib and LY3214496, are also in clinical development and may be of benefit to individuals with RASopathies (Liu, Yang, Geng, & Huang, 2018). Because ERK activation is noted in most RASopathy models, ERK inhibitors merit testing in RASopathy models of diverse genotypes.

6 |. LESSONS LEARNED FROM INITIAL COMPASSIONATE USE: CONSIDERATIONS FOR NEW TREATMENTS

As investigators, affected families, and biopharmaceutical companies coalesce around the exciting development of mechanistic therapeutics for the RASopathies, be those through drug repurposing or novel drug development, there are several issues and concepts that might be useful to consider, which will be addressed here.

As the RASopathies are traits that affect multiple organs and tissues, a critical issue is determining the best therapeutic target(s). As described above, the activity in repurposing MEKis has focused on severe manifestations for which conventional approaches are either non-existent or highly morbid (e.g., severe HCM in infants, severe lymphovascular dysfunction). While rational, this has the downside of applicability to a limited percentage of affected individuals, which may not result in maximal utility and may present barriers to successful completion of clinical trials. On the other hand, targeting a common issue in the RASopathies—consider short stature as an example—would potentially benefit more affected individuals but might raise greater concerns about safety, both due to the relative health impact of that issue and the availability of an FDA-approved therapy. A potential solution to this dilemma is to try a novel drug in a severe, rare aspect of the RASopathies and use the insights about efficacy and adverse events from that experience to determine the advisability of subsequent trials for commoner, less morbid issues.

Precedent from animal studies is clearly in favor of such an approach since several extracardiac phenotypes of RASopathies have been ameliorated using targeted approaches in mouse models. Examples include attenuation of the craniofacial phenotype and improved growth in the RAF1 L613V mouse model when MEKi is started within the first 4 weeks of life (Wu et al., 2011). In a Drosophila model of gain-of-function mutations of PTPN11, neuronal development is normal, but homeostasis is altered (Lee et al., 2014). Blocking SHP-2 resolved these issues. Mouse models for the common PTPN11 mutations N308D and D61G showed hippocampal-dependent spatial learning impairments and deficits in synaptic strengths (Lee et al., 2014). Treatment with the MEKi SL327 and the HMG-CoA reductase inhibitor lovastatin, which also inhibits prenylation and thus RAS membrane association, improved synaptic strength and learning in these two models. While often not classified as a RASopathy per se, mutations in SYNGAP1 are a frequent cause of non-syndromic developmental delay through hyperphosphorylation of the MEK and ERK, with ensuing synaptic dysfunction. Treatment of a mouse model of SYNGAP1 deficiency with the MEKi PD-0325901 resulted in normalization of basal synaptic activity, but their long-term potentiation implicated in learning (Kopanitsa et al., 2018). These results point to the possibility that specific phenotypes of SYNGAP1 deficiency may be improved through treatment with MEKi.

A related issue for clinical trials will be inclusion criteria with respect to RASopathy genotype. Consider this thought experiment: there is evidence from pre-clinical studies to suggest that mechanistic therapy might improve neurocognitive function in the RASopathies. With a candidate therapy in hand, would one undertake a clinical trial with patient with genotypes associated with more severe neurocognitive impairments (i.e., Costello and CFC syndromes) or mild impairments (i.e., NS)? Relatedly, as mechanistic therapy advances beyond the most severe, early-onset forms of HCM, which are almost entirely observed in babies with NS and NSML, would all RASopathy-associated HCM be eligible? Can a prospective randomized trial be justified at all if additional data from compassionate care emerge and underpin the case for strong beneficial effects of MEKi/mTORi? Clearly, issues of equity and non-discrimination will come to the fore, and the community will need to carefully avoid decisions arising from unintentional biases. Innovative strategies for trial design in small populations are now available (Friede et al., 2018; Hee et al., 2017; Ursino & Stallard, 2021). One example is delayed start of treatment in one group versus the other, and comparison of patient groups according to the time point at which treatment was started. This could help avoid a situation in which patients on a placebo arm are denied the potential benefits of MEKi/mTORi.

In considering repurposing mechanistic therapies for the RASopathies, there are a number of important considerations. For repurposed drugs, there will generally be prior experience in one or more other clinical contexts. For the MEKis, that has meant use in patients with cancer, often adults. Aside from the obvious potential differences in drug metabolism based on age, there is another major dividing line: patients with cancer have gain of function for the RAS/MAPK pathway only in their cancer due to a somatic mutation, whereas patients with RASopathies have germline variants. Also of note, modeling of CFC-associated MEK variants in fruit fly embryos has shown that the complexities of the regulation of signal transduction pathways results in gain-of-function alleles engendering increased signaling in some tissues but decreased signaling in others (Goyal et al., 2017). This may be due to feedback loops in which some activating variants may lead to strong negative feedback resulting in reduced MAPK signaling in certain other tissues. Taken together, predicting the side-effect profile of any drug targeting RAS signaling developed in patients with cancers may or may not prove accurate for the RASopathy context. Optimistically, some drug side effects may never occur in patients with RASopathies. Realistically, patients with RASopathies will experience some of those side effects as already apparent from the dermatological issues observed in patients with NS treated with MEKis and could even encounter novel ones never observed in older patients with somatic RAS pathway mutations.

Two additional considerations might also impact the side effects associated with mechanistic therapies for the RASopathies: dosing and treatment duration. For dosing, the comparison to cancer treatment is germane. To be effective, the goal in oncologic therapeutics is to shut down RAS signaling in order to induce apoptosis in cancer tissues that are “RAS addicted.” The therapeutic goal in the RASopathies is quite different: to normalize RAS signaling levels. For repurposing therapeutics that directly target the canonical RAS/MAPK pathway, that might be achieved through dose reduction (as has been done with MEKis to date, described above). However, this concept also suggests that therapeutics that alter the RAS/MAPK signaling network, perhaps targeting more peripheral or regulatory proteins, which would not be strategically useful for cancer, might be effective for the RASopathies. If such an approach also had fewer side effects, it might facilitate use for non-life-threatening aspects of the RASopathies.

Another substantive difference between mechanistic RAS/MAPK therapy in cancer versus the RASopathies is treatment duration. For cancer, treatment, generally undertaken with the maximally tolerated dosing, is relatively brief, consistent with the goal of inducing apoptosis in the tumor and necessitated by toxicities. Treatment duration in the RASopathies will almost certainly need to be longer than for cancer and will likely vary with the indication. This issue has already arisen with the use of MEKis for NS-associated HCM as described above. Trial and error may be needed to determine the appropriate duration for this indication. On the other hand, hypothetically effective treatment for neurocognitive dysfunction would most likely necessitate life-long use as the therapeutic purpose is to restore normal neuronal homeostasis, something that would be lost shortly after stopping an effective medicine.

In summary, the RASopathy community stands on the brink of developing meaningful mechanistic therapies for various aspects of these traits. As discussed here, there are several thorny issues to consider and questions to be asked as we proceed. By being deliberate, we should be able to reach wide consensus and avoid many of the potential pitfalls.

ACKNOWLEDGMENTS

Bruce D. Gelb was supported by a grant from the United States’ National Institutes of Health (HL135742). Marielle E. Yohe was supported by the National Cancer Institute intramural program. Marielle E. Yohe was supported by the National Cancer Institute intramural program. Cordula Wolf is member of the European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-Heart). Gregor Andelfinger was supported by the Banque Nationale Research Excellence Chair in Cardiovascular Genetics.

Funding information

Banque Nationale Research Excellence Chair in Cardiovascular Genetics; Fondation Sainte Justine; Million Dollar Bike Ride; National Institutes of Health

Footnotes

DISCLOSURE STATEMENT

Bruce D. Gelb: Dr. Bruce Gelb is a named inventor on issued patents related to PTPN11, SHOC2, RAF1, and SOS1 mutations. Icahn School of Medicine at Mount Sinai licensed the patent to several diagnostics companies (Correlegan, GeneDx, LabCorp, and Prevention Genetics) and has received royalty payments, some of which are distributed to Dr. Gelb. Dr. Gelb is the recipient of a Sponsored Research Agreement from Onconova and Day One Biopharmaceuticals. Dr. Gelb is a consultant for Day One Biopharmaceuticals, Inc. and BioMarin Pharmaceuticals, Inc. Marielle E. Yohe: No disclosure. Cordula Wolf: Honoraria: Novo Nordisk; Dr. Wolf is a consultant for Day One Bio-pharmaceuticals, Inc., BioMarin Pharmaceuticals, Adrenomed AG, and Pliant Therapeutics. Gregor Andelfinger: Dr. Andelfinger is a consultant for Day One Biopharmaceuticals, Inc, and BioMarin Pharmaceuticals.

DATA AVAILABILITY STATEMENT

This manuscript does not contain data that would require public availability.

REFERENCES

  1. Adamopoulos C, Ahmed TA, Tucker MR, Ung PMU, Xiao M, Karoulia Z, … Poulikakos PI (2021). Exploiting allosteric properties of RAF and MEK inhibitors to target therapy-resistant tumors driven by oncogenic BRAF signaling. Cancer Discovery, 11(7), 1716–1735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Andelfinger G, Marquis C, Raboisson MJ, Theoret Y, Waldmuller S, Wiegand G, … Hofbeck M (2019). Hypertrophic cardiomyopathy in Noonan syndrome treated by MEK-inhibition. Journal of the American College of Cardiology, 73(17), 2237–2239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Baldari S, Di Rocco G, & Toietta G (2020). Current biomedical use of copper chelation therapy. International Journal of Molecular Sciences, 21(3), 1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Baranyi M, Buday L, & Hegedus B (2020). K-Ras prenylation as a potential anticancer target. Cancer Metastasis Reviews, 39(4), 1127–1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Blumenschein GR Jr., Smit EF, Planchard D, Kim DW, Cadranel J, De Pas T, … Janne PA (2015). A randomized phase II study of the MEK1/MEK2 inhibitor trametinib (GSK1120212) compared with docetaxel in KRAS-mutant advanced non-small-cell lung cancer (NSCLC) dagger. Annals of Oncology, 26(5), 894–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bobone S, Pannone L, Biondi B, Solman M, Flex E, Canale VC, … Stella L (2021). Targeting oncogenic Src homology 2 domain-containing phosphatase 2 (SHP2) by inhibiting its protein–protein interactions. Journal of Medicinal Chemistry, 64(21), 15973–15990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Brady DC, Crowe MS, Turski ML, Hobbs GA, Yao X, Chaikuad A, … Counter CM (2014). Copper is required for oncogenic BRAF signalling and tumorigenesis. Nature, 509(7501), 492–496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Calcagni G, Adorisio R, Martinelli S, Grutter G, Baban A, Versacci P, … Marino B (2018). Clinical presentation and natural history of hypertrophic cardiomyopathy in RASopathies. Heart Failure Clinics, 14(2), 225–235. [DOI] [PubMed] [Google Scholar]
  9. Casey D, Demko S, Sinha A, Mishra-Kalyani PS, Shen YL, Khasar S, … Singh H (2021). FDA approval summary: Selumetinib for plexiform neurofibroma. Clinical Cancer Research, 27(15), 4142–4146. [DOI] [PubMed] [Google Scholar]
  10. Castel P, Cheng A, Cuevas-Navarro A, Everman DB, Papageorge AG, Simanshu DK, … McCormick F (2019). RIT1 oncoproteins escape LZTR1-mediated proteolysis. Science, 363(6432), 1226–1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Chakraborty A, Beasley G, Martinez H, Jesudas R, Anton-Martin P, Christakopoulos G, & Kramer J (2022). Selumetinib for refractory pulmonary and gastrointestinal bleeding in Noonan syndrome. Pediatrics, 150(4), 1–5. [DOI] [PubMed] [Google Scholar]
  12. Chen S, Chen L, Jiang Y, Xu H, Sun Y, Shi H, … Yan J (2022). Early outcomes of septal myectomy for obstructive hypertrophic cardiomyopathy in children with Noonan syndrome. Seminars in Thoracic and Cardiovascular Surgery, 34(2), 655–665. [DOI] [PubMed] [Google Scholar]
  13. Chen Y, & Zhou X (2020). Research progress of mTOR inhibitors. European Journal of Medicinal Chemistry, 208, 112820. [DOI] [PubMed] [Google Scholar]
  14. Cleary JM, Wang V, Heist RS, Kopetz ES, Mitchell EP, Zwiebel JA, … Flaherty KT (2021). Differential outcomes in codon 12/13 and codon 61 NRAS-mutated cancers in the phase II NCI-MATCH trial of binimetinib in patients with NRAS-mutated tumors. Clinical Cancer Research, 27(11), 2996–3004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Cox AD, Der CJ, & Philips MR (2015). Targeting RAS membrane association: Back to the future for anti-RAS drug discovery? Clinical Cancer Research, 21(8), 1819–1827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Cox TP, Vance CJ, Daley SK, Papendieck C, McGregor H, Kuo P, & Witte MH (2022). Systematic literature review of lymphatic imaging-guided procedural management of Noonan syndrome. Journal of Vascular Surgery. Venous and Lymphatic Disorders, 10(5), 1192–1196 e1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Crooke ST, Baker BF, Crooke RM, & Liang XH (2021). Antisense technology: An overview and prospectus. Nature Reviews. Drug Discovery, 20(6), 427–453. [DOI] [PubMed] [Google Scholar]
  18. Davies BR, Greenwood H, Dudley P, Crafter C, Yu DH, Zhang J, … Pass M (2012). Preclinical pharmacology of AZD5363, an inhibitor of AKT: Pharmacodynamics, antitumor activity, and correlation of monotherapy activity with genetic background. Molecular Cancer Therapeutics, 11(4), 873–887. [DOI] [PubMed] [Google Scholar]
  19. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, … Futreal PA (2002). Mutations of the BRAF gene in human cancer. Nature, 417(6892), 949–954. [DOI] [PubMed] [Google Scholar]
  20. de Blank PMK, Gross AM, Akshintala S, Blakeley JO, Bollag G, Cannon A, … Fisher MJ (2022). MEK inhibitors for neurofibromatosis type 1 manifestations: Clinical evidence and consensus. Neuro-Oncology, 24(11), 1845–1856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Dori Y, Smith C, Pinto E, Snyder K, March ME, Hakonarson H, & Belasco J (2020). Severe lymphatic disorder resolved with MEK inhibition in a patient with Noonan syndrome and SOS1 mutation. Pediatrics, 146(6), 1–5. [DOI] [PubMed] [Google Scholar]
  22. Dummer R, Ascierto PA, Gogas HJ, Arance A, Mandala M, Liszkay G, … Flaherty KT (2018). Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): A multicentre, open-label, randomised phase 3 trial. The Lancet Oncology, 19(5), 603–615. [DOI] [PubMed] [Google Scholar]
  23. Dummer R, Schadendorf D, Ascierto PA, Arance A, Dutriaux C, Di Giacomo AM, … Flaherty K (2017). Binimetinib versus dacarbazine in patients with advanced NRAS-mutant melanoma (NEMO): A multi-centre, open-label, randomised, phase 3 trial. The Lancet Oncology, 18(4), 435–445. [DOI] [PubMed] [Google Scholar]
  24. Durrant DE, & Morrison DK (2018). Targeting the Raf kinases in human cancer: The Raf dimer dilemma. British Journal of Cancer, 118(1), 3–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. End DW, Smets G, Todd AV, Applegate TL, Fuery CJ, Angibaud P, … Bowden C (2001). Characterization of the antitumor effects of the selective farnesyl protein transferase inhibitor R115777 in vivo and in vitro. Cancer Research, 61(1), 131–137. [PubMed] [Google Scholar]
  26. Forde K, Resta N, Ranieri C, Rea D, Kubassova O, Hinton M, … Dvorakova V (2021). Clinical experience with the AKT1 inhibitor miransertib in two children with PIK3CA-related overgrowth syndrome. Orphanet Journal of Rare Diseases, 16(1), 109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Friede T, Posch M, Zohar S, Alberti C, Benda N, Comets E, … Stallard N (2018). Recent advances in methodology for clinical trials in small populations: The InSPiRe project. Orphanet Journal of Rare Diseases, 13(1), 186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Fritsch C, Huang A, Chatenay-Rivauday C, Schnell C, Reddy A, Liu M, … Sellers WR (2014). Characterization of the novel and specific PI3Kalpha inhibitor NVP-BYL719 and development of the patient stratification strategy for clinical trials. Molecular Cancer Therapeutics, 13(5), 1117–1129. [DOI] [PubMed] [Google Scholar]
  29. Germann UA, Furey BF, Markland W, Hoover RR, Aronov AM, Roix JJ, … Welsch DJ (2017). Targeting the MAPK signaling pathway in cancer: Promising preclinical activity with the novel selective ERK1/2 inhibitor BVD-523 (Ulixertinib). Molecular Cancer Therapeutics, 16(11), 2351–2363. [DOI] [PubMed] [Google Scholar]
  30. Goyal Y, Jindal GA, Pelliccia JL, Yamaya K, Yeung E, Futran AS, … Shvartsman SY (2017). Divergent effects of intrinsically active MEK variants on developmental Ras signaling. Nature Genetics, 49(3), 465–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Gross AM, Wolters PL, Dombi E, Baldwin A, Whitcomb P, Fisher MJ, … Widemann BC (2020). Selumetinib in children with inoperable plexiform neurofibromas. The New England Journal of Medicine, 382(15), 1430–1442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Hahn A, Lauriol J, Thul J, Behnke-Hall K, Logeswaran T, Schanzer A, … Schranz D (2015). Rapidly progressive hypertrophic cardiomyopathy in an infant with Noonan syndrome with multiple lentigines: Palliative treatment with a rapamycin analog. American Journal of Medical Genetics Part A, 167A(4), 744–751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Hartung AM, Swensen J, Uriz IE, Lapin M, Kristjansdottir K, Petersen US, … Andresen BS (2016). The splicing efficiency of activating HRAS mutations can determine Costello syndrome phenotype and frequency in cancer. PLoS Genetics, 12(5), e1006039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Hebron KE, Hernandez ER, & Yohe ME (2022). The RASopathies: From pathogenetics to therapeutics. Disease Models & Mechanisms, 15(2), 1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Hee SW, Willis A, Tudur Smith C, Day S, Miller F, Madan J, … Stallard N (2017). Does the low prevalence affect the sample size of interventional clinical trials of rare diseases? An analysis of data from the aggregate analysis of clinicaltrials.gov. Orphanet Journal of Rare Diseases, 12(1), 44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Hemmati P, Dearani JA, Daly RC, King KS, Ammash NM, Cetta F, & Schaff HV (2019). Early outcomes of cardiac surgery in patients with Noonan syndrome. Seminars in Thoracic and Cardiovascular Surgery, 31(3), 507–513. [DOI] [PubMed] [Google Scholar]
  37. Hernandez-Porras I, Fabbiano S, Schuhmacher AJ, Aicher A, Canamero M, Camara JA, … Barbacid M (2014). K-RasV14I recapitulates Noonan syndrome in mice. Proceedings of the National Academy of Sciences of the United States of America, 111(46), 16395–16400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Hickey EJ, Mehta R, Elmi M, Asoh K, McCrindle BW, Williams WG, … Benson L (2011). Survival implications: Hypertrophic cardiomyopathy in Noonan syndrome. Congenital Heart Disease, 6(1), 41–47. [DOI] [PubMed] [Google Scholar]
  39. Hillig RC, Sautier B, Schroeder J, Moosmayer D, Hilpmann A, Stegmann CM, … Bader B (2019). Discovery of potent SOS1 inhibitors that block RAS activation via disruption of the RAS-SOS1 interaction. Proceedings of the National Academy of Sciences of the United States of America, 116(7), 2551–2560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Hong DS, Fakih MG, Strickler JH, Desai J, Durm GA, Shapiro GI, … Li BT (2020). KRAS(G12C) inhibition with sotorasib in advanced solid tumors. The New England Journal of Medicine, 383(13), 1207–1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Jaffre F, Miller CL, Schanzer A, Evans T, Roberts AE, Hahn A, & Kontaridis MI (2019). Inducible pluripotent stem cell-derived cardiomyocytes reveal aberrant extracellular regulated kinase 5 and mitogen-activated protein kinase kinase 1/2 signaling concomitantly promote hypertrophic cardiomyopathy in RAF1-associated Noonan syndrome. Circulation, 140(3), 207–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Janku F, Iyer G, Spreafico A, Yamamoto N, Bang Y-J, Elez E, … Dummer R (2018). A phase I study of LXH254 in patients (pts) with advanced solid tumors harboring MAPK pathway alterations. Journal of Clinical Oncology, 36(15_suppl), 2586. [Google Scholar]
  43. Josowitz R, Mulero-Navarro S, Rodriguez NA, Falce C, Cohen N, Ullian EM, … Gelb BD (2016). Autonomous and non-autonomous defects underlie hypertrophic cardiomyopathy in BRAF-mutant hiPSC-derived cardiomyocytes. Stem Cell Reports, 7(3), 355–369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Kamerkar S, LeBleu VS, Sugimoto H, Yang S, Ruivo CF, Melo SA, … Kalluri R (2017). Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature, 546(7659), 498–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Kenney C, Kunst T, Webb S, Christina D Jr., Arrowood C, Steinberg SM, … Rudloff U (2021). Phase II study of selumetinib, an orally active inhibitor of MEK1 and MEK2 kinases, in KRAS(G12R)-mutant pancreatic ductal adenocarcinoma. Investigational New Drugs, 39(3), 821–828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Kessler D, Bergner A, Bottcher J, Fischer G, Dobel S, Hinkel M, … McConnell DB (2020). Drugging all RAS isoforms with one pocket. Future Medicinal Chemistry, 12(21), 1911–1923. [DOI] [PubMed] [Google Scholar]
  47. Kessler D, Gmachl M, Mantoulidis A, Martin LJ, Zoephel A, Mayer M, … McConnell DB (2019). Drugging an undruggable pocket on KRAS. Proceedings of the National Academy of Sciences of the United States of America, 116(32), 15823–15829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Klesse LJ, Jordan JT, Radtke HB, Rosser T, Schorry E, Ullrich N, … Yohay K (2020). The use of MEK inhibitors in neurofibromatosis type 1-associated tumors and management of toxicities. The Oncologist, 25(7), e1109–e1116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Kobialka P, Sabata H, Vilalta O, Gouveia L, Angulo-Urarte A, Muixi L, … Graupera M (2022). The onset of PI3K-related vascular malformations occurs during angiogenesis and is prevented by the AKT inhibitor miransertib. EMBO Molecular Medicine, 14(7), e15619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Koltun ES, Rice MA, Gustafson C, Wilds D, Jiang J, Lee BJ, … Sing M (2022). Direct targeting of KRASG12X mutant cancers with RMC-6236, a first-in-class, RAS-selective, orally bioavailable, tri-complex RASMULTI(ON) inhibitor. Cancer Research, 82 Abstract 3597. [Google Scholar]
  51. Kondyli M, Larouche V, Saint-Martin C, Ellezam B, Pouliot L, Sinnett D, … Perreault S (2018). Trametinib for progressive pediatric low-grade gliomas. Journal of Neuro-Oncology, 140(2), 435–444. [DOI] [PubMed] [Google Scholar]
  52. Kopanitsa MV, Gou G, Afinowi NO, Bayes A, Grant SGN, & Komiyama NH (2018). Chronic treatment with a MEK inhibitor reverses enhanced excitatory field potentials in Syngap1(+/−) mice. Pharmacological Reports, 70(4), 777–783. [DOI] [PubMed] [Google Scholar]
  53. Krab LC, de Goede-Bolder A, Aarsen FK, Pluijm SM, Bouman MJ, van der Geest JN, … Elgersma Y (2008). Effect of simvastatin on cognitive functioning in children with neurofibromatosis type 1: A randomized controlled trial. JAMA, 300(3), 287–294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Kratz CP, Franke L, Peters H, Kohlschmidt N, Kazmierczak B, Finckh U, … Zenker M (2015). Cancer spectrum and frequency among children with Noonan, Costello, and cardio-facio-cutaneous syndromes. British Journal of Cancer, 112(8), 1392–1397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Krause G, Hassenruck F, & Hallek M (2018). Copanlisib for treatment of B-cell malignancies: The development of a PI3K inhibitor with considerable differences to idelalisib. Drug Design, Development and Therapy, 12, 2577–2590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Lai LP, Fer N, Burgan W, Wall VE, Xu B, Soppet D, … McCormick F (2022). Classical RAS proteins are not essential for paradoxical ERK activation induced by RAF inhibitors. Proceedings of the National Academy of Sciences of the United States of America, 119(5), 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. LaMarche MJ, Acker M, Argintaru A, Bauer D, Boisclair J, Chan H, … Zhu S (2020). Identification of TNO155, an allosteric SHP2 inhibitor for the treatment of cancer. Journal of Medicinal Chemistry, 63(22), 13578–13594. [DOI] [PubMed] [Google Scholar]
  58. Lannutti BJ, Meadows SA, Herman SE, Kashishian A, Steiner B, Johnson AJ, … Giese NA (2011). CAL-101, a p110delta selective phosphatidylinositol-3-kinase inhibitor for the treatment of B-cell malignancies, inhibits PI3K signaling and cellular viability. Blood, 117(2), 591–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Laredo M, Khraiche D, Raisky O, Gaudin R, Bajolle F, Maltret A, … Vouhe PR (2018). Long-term results of the modified Konno procedure in high-risk children with obstructive hypertrophic cardiomyopathy. The Journal of Thoracic and Cardiovascular Surgery, 156(6), 2285–2294 e2282. [DOI] [PubMed] [Google Scholar]
  60. Larkin J, Ascierto PA, Dreno B, Atkinson V, Liszkay G, Maio M, … Ribas A (2014). Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. The New England Journal of Medicine, 371(20), 1867–1876. [DOI] [PubMed] [Google Scholar]
  61. Lee YS, Ehninger D, Zhou M, Oh JY, Kang M, Kwak C, … Silva AJ (2014). Mechanism and treatment for learning and memory deficits in mouse models of Noonan syndrome. Nature Neuroscience, 17(12), 1736–1743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Leoni C, Gullo G, Resta N, Fagotti A, Onesimo R, Schwartz B, … Zampino G (2019). First evidence of a therapeutic effect of miransertib in a teenager with Proteus syndrome and ovarian carcinoma. American Journal of Medical Genetics Part A, 179(7), 1319–1324. [DOI] [PubMed] [Google Scholar]
  63. Li D, March ME, Gutierrez-Uzquiza A, Kao C, Seiler C, Pinto E, … Hakonarson H (2019). ARAF recurrent mutation causes central conducting lymphatic anomaly treatable with a MEK inhibitor. Nature Medicine, 25(7), 1116–1122. [DOI] [PubMed] [Google Scholar]
  64. Li W, Cui Y, Kushner SA, Brown RA, Jentsch JD, Frankland PW, … Silva AJ (2005). The HMG-CoA reductase inhibitor lovastatin reverses the learning and attention deficits in a mouse model of neurofibromatosis type 1. Current Biology, 15(21), 1961–1967. [DOI] [PubMed] [Google Scholar]
  65. Lim S, Boyer N, Boo N, Huang C, Venkatachalam G, Angela Juang YC, … Partridge AW (2021). Discovery of cell active macrocyclic peptides with on-target inhibition of KRAS signaling. Chemical Science, 12(48), 15975–15987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Lin J, Sampath D, Nannini MA, Lee BB, Degtyarev M, Oeh J, … Lin K (2013). Targeting activated Akt with GDC-0068, a novel selective Akt inhibitor that is efficacious in multiple tumor models. Clinical Cancer Research, 19(7), 1760–1772. [DOI] [PubMed] [Google Scholar]
  67. Lioncino M, Fusco A, Monda E, Colonna D, Sibilio M, Caiazza M, … Limongelli G (2022). Severe lymphatic disorder and multifocal atrial tachycardia treated with trametinib in a patient with Noonan syndrome and SOS1 mutation. Genes, 13(9), 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Liu F, Yang X, Geng M, & Huang M (2018). Targeting ERK, an Achilles’ heel of the MAPK pathway, in cancer therapy. Acta Pharmaceutica Sinica B, 8(4), 552–562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Luckman SP, Hughes DE, Coxon FP, Graham R, Russell G, & Rogers MJ (1998). Nitrogen-containing bisphosphonates inhibit the mevalonate pathway and prevent post-translational prenylation of GTP-binding proteins, including Ras. Journal of Bone and Mineral Research, 13(4), 581–589. [DOI] [PubMed] [Google Scholar]
  70. Marin TM, Keith K, Davies B, Conner DA, Guha P, Kalaitzidis D, … Kontaridis MI (2011). Rapamycin reverses hypertrophic cardiomyopathy in a mouse model of LEOPARD syndrome-associated PTPN11 mutation. The Journal of Clinical Investigation, 121(3), 1026–1043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. McCallen LM, Ameduri RK, Denfield SW, Dodd DA, Everitt MD, Johnson JN, … Chatfield KC (2019). Cardiac transplantation in children with Noonan syndrome. Pediatric Transplantation, 23(6), e13535. [DOI] [PubMed] [Google Scholar]
  72. Meisner JK, Bradley DJ, & Russell MW (2021). Molecular management of multifocal atrial tachycardia in Noonan’s syndrome with MEK1/2 inhibitor trametinib. Circulation: Genomic and Precision Medicine, 14(5), e003327. [DOI] [PubMed] [Google Scholar]
  73. Monaco KA, Delach S, Yuan J, Mishina Y, Fordjour P, Labrot E, … Caponigro G (2021). LXH254, a potent and selective ARAF-sparing inhibitor of BRAF and CRAF for the treatment of MAPK-driven tumors. Clinical Cancer Research, 27(7), 2061–2073. [DOI] [PubMed] [Google Scholar]
  74. Moore AR, Rosenberg SC, McCormick F, & Malek S (2020). RAS-targeted therapies: Is the undruggable drugged? Nature Reviews. Drug Discovery, 19(8), 533–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Morrison DK (2012). MAP kinase pathways. Cold Spring Harbor Perspectives in Biology, 4(11), 1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Mussa A, Carli D, Giorgio E, Villar AM, Cardaropoli S, Carbonara C, … Ferrero GB (2021). MEK inhibition in a newborn with RAF1-associated Noonan syndrome ameliorates hypertrophic cardio-myopathy but is insufficient to revert pulmonary vascular disease. Genes, 13(1), 1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Nagasaka M, Li Y, Sukari A, Ou SI, Al-Hallak MN, & Azmi AS (2020). KRAS G12C game of thrones, which direct KRAS inhibitor will claim the iron throne? Cancer Treatment Reviews, 84, 101974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Nakamura T, Gulick J, Pratt R, & Robbins J (2009). Noonan syndrome is associated with enhanced pERK activity, the repression of which can prevent craniofacial malformations. Proceedings of the National Academy of Sciences of the United States of America, 106(36), 15436–15441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Nakano TA, Rankin AW, Annam A, Kulungowski AM, McCallen LM, Hill LR, & Chatfield KC (2022). Trametinib for refractory chylous effusions and systemic complications in children with Noonan syndrome. The Journal of Pediatrics, 248, 81–88.e1. [DOI] [PubMed] [Google Scholar]
  80. Nichols RJ, Haderk F, Stahlhut C, Schulze CJ, Hemmati G, Wildes D, … Bivona TG (2018). RAS nucleotide cycling underlies the SHP2 phosphatase dependence of mutant BRAF-, NF1- and RAS-driven cancers. Nature Cell Biology, 20(9), 1064–1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Nichols RJ, Yang YC, Cregg JM, Schulze CJ, Wang Z, Dua R, … Smith JAM (2022). RMC-6291, a next-generation tri-complex KRASG12C(ON) inhibitor, outperforms KRASG12C(OFF) inhibitors in preclinical models of KRASG12C cancers. Cancer Research, 82-(12_Suppl) Abstract 3595. [Google Scholar]
  82. Oba D, Inoue SI, Miyagawa-Tomita S, Nakashima Y, Niihori T, Yamaguchi S, … Aoki Y (2018). Mice with an oncogenic HRAS mutation are resistant to high-fat diet-induced obesity and exhibit impaired hepatic energy homeostasis. eBioMedicine, 27, 138–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Ostman-Smith I (2014). Beta-blockers in pediatric hypertrophic cardiomyopathies. Reviews on Recent Clinical Trials, 9(2), 82–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Ours CA, Sapp JC, Hodges MB, de Moya AJ, & Biesecker LG (2021). Case report: Five-year experience of AKT inhibition with miransertib (MK-7075) in an individual with Proteus syndrome. Cold Spring Harbor Molecular Case Studies, 7(6), a006134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Padua RAP, Sun Y, Marko I, Pitsawong W, Stiller JB, Otten R, & Kern D (2018). Mechanism of activating mutations and allosteric drug inhibition of the phosphatase SHP2. Nature Communications, 9(1), 4507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Parikh K, Banna G, Liu SV, Friedlaender A, Desai A, Subbiah V, & Addeo A (2022). Drugging KRAS: Current perspectives and state-of-art review. Journal of Hematology & Oncology, 15(1), 152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Payne JM, Barton B, Ullrich NJ, Cantor A, Hearps SJ, Cutter G, … Consortium NFCT. (2016). Randomized placebo-controlled study of lovastatin in children with neurofibromatosis type 1. Neurology, 87(24), 2575–2584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Peng SB, Henry JR, Kaufman MD, Lu WP, Smith BD, Vogeti S, … Flynn DL (2015). Inhibition of RAF isoforms and active dimers by LY3009120 leads to anti-tumor activities in RAS or BRAF mutant cancers. Cancer Cell, 28(3), 384–398. [DOI] [PubMed] [Google Scholar]
  89. Poterucha JT, Johnson JN, O’Leary PW, Connolly HM, Niaz T, Maleszewski JJ, … Eidem BW (2015). Surgical ventricular septal myectomy for patients with Noonan syndrome and symptomatic left ventricular outflow tract obstruction. The American Journal of Cardiology, 116(7), 1116–1121. [DOI] [PubMed] [Google Scholar]
  90. Prior IA, Hood FE, & Hartley JL (2020). The frequency of Ras mutations in cancer. Cancer Research, 80(14), 2969–2974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Ramurthy S, Taft BR, Aversa RJ, Barsanti PA, Burger MT, Lou Y, … Stuart DD (2020). Design and discovery of N-(3-(2-(2-hydroxyethoxy)-6-morpholinopyridin-4-yl)-4-methylphenyl)-2-(trifluorom ethyl)isonicotinamide, a selective, efficacious, and well-tolerated RAF inhibitor targeting RAS mutant cancers: The path to the clinic. Journal of Medicinal Chemistry, 63(5), 2013–2027. [DOI] [PubMed] [Google Scholar]
  92. Robert C, Karaszewska B, Schachter J, Rutkowski P, Mackiewicz A, Stroiakovski D, … Schadendorf D (2015). Improved overall survival in melanoma with combined dabrafenib and trametinib. The New England Journal of Medicine, 372(1), 30–39. [DOI] [PubMed] [Google Scholar]
  93. Roberts AE, Allanson JE, Tartaglia M, & Gelb BD (2013). Noonan syndrome. Lancet, 381(9863), 333–342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Rodriguez-Viciana P, Tetsu O, Tidyman WE, Estep AL, Conger BA, Cruz MS, … Rauen KA (2006). Germline mutations in genes within the MAPK pathway cause cardio-facio-cutaneous syndrome. Science, 311(5765), 1287–1290. [DOI] [PubMed] [Google Scholar]
  95. Roskoski R Jr. (2018). Targeting oncogenic Raf protein-serine/threonine kinases in human cancers. Pharmacological Research, 135, 239–258. [DOI] [PubMed] [Google Scholar]
  96. Ross SJ, Revenko AS, Hanson LL, Ellston R, Staniszewska A, Whalley N, … Macleod AR (2017). Targeting KRAS-dependent tumors with AZD4785, a high-affinity therapeutic antisense oligonucleotide inhibitor of KRAS. Science Translational Medicine, 9(394), 1–13. [DOI] [PubMed] [Google Scholar]
  97. Schreiber J, Grimbergen LA, Overwater I, Vaart TV, Stedehouder J, Schuhmacher AJ, … Elgersma Y (2017). Mechanisms underlying cognitive deficits in a mouse model for Costello syndrome are distinct from other RASopathy mouse models. Scientific Reports, 7(1), 1256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Sebti SM, & Hamilton AD (2000). Farnesyltransferase and geranylger-anyltransferase I inhibitors in cancer therapy: Important mechanistic and bench to bedside issues. Expert Opinion on Investigational Drugs, 9(12), 2767–2782. [DOI] [PubMed] [Google Scholar]
  99. Senawong T, Phuchareon J, Ohara O, McCormick F, Rauen KA, & Tetsu O (2008). Germline mutations of MEK in cardio-faciocutaneous syndrome are sensitive to MEK and RAF inhibition: Implications for therapeutic options. Human Molecular Genetics, 17(3), 419–430. [DOI] [PubMed] [Google Scholar]
  100. Sepp-Lorenzino L, Ma Z, Rands E, Kohl NE, Gibbs JB, Oliff A, & Rosen N (1995). A peptidomimetic inhibitor of farnesyl:protein transferase blocks the anchorage-dependent and -independent growth of human tumor cell lines. Cancer Research, 55(22), 5302–5309. [PubMed] [Google Scholar]
  101. Sideris E, Tng ETV, & Chee P (2021). Lymphatic malformation responsive to sirolimus in keratinocytic epidermal nevus syndrome with KRAS mutation: A case and brief literature discussion. Case Reports in Dermatology, 13(1), 195–201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Sleutjes J, Kleimeier L, Leenders E, Klein W, & Draaisma J (2022). Lymphatic abnormalities in Noonan syndrome spectrum disorders: A systematic review. Molecular Syndromology, 13(1), 1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Smith MC, Mader MM, Cook JA, Iversen P, Ajamie R, Perkins E, … Wacheck V (2016). Characterization of LY3023414, a novel PI3K/mTOR dual inhibitor eliciting transient target modulation to impede tumor growth. Molecular Cancer Therapeutics, 15(10), 2344–2356. [DOI] [PubMed] [Google Scholar]
  104. Sparks TN, Lianoglou BR, Adami RR, Pluym ID, Holliman K, Duffy J, … University of California Fetal-Maternal C, University of California SFCfM-FPM. (2020). Exome sequencing for prenatal diagnosis in nonimmune hydrops fetalis. The New England Journal of Medicine, 383(18), 1746–1756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Sullivan RJ, Hollebecque A, Flaherty KT, Shapiro GI, Rodon Ahnert J, Millward MJ, … Hong DS (2020). A phase I study of LY3009120, a Pan-RAF inhibitor, in patients with advanced or metastatic cancer. Molecular Cancer Therapeutics, 19(2), 460–467. [DOI] [PubMed] [Google Scholar]
  106. Sun Y, Alberta JA, Pilarz C, Calligaris D, Chadwick EJ, Ramkissoon SH, … Stiles CD (2017). A brain-penetrant RAF dimer antagonist for the noncanonical BRAF oncoprotein of pediatric low-grade astrocytomas. Neuro-Oncology, 19(6), 774–785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Swarts JW, Kleimeier LER, Leenders E, Rinne T, Klein WM, & Draaisma JMT (2022). Lymphatic anomalies during lifetime in patients with Noonan syndrome: Retrospective cohort study. American Journal of Medical Genetics Part A, 188(11), 3242–3261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Takahara S, Inoue SI, Miyagawa-Tomita S, Matsuura K, Nakashima Y, Niihori T, … Aoki Y (2019). New Noonan syndrome model mice with RIT1 mutation exhibit cardiac hypertrophy and susceptibility to beta-adrenergic stimulation-induced cardiac fibrosis. eBioMedicine, 42, 43–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Trippett T, Toledano H, Campbell Hewson Q, Verschuur A, Langevin AM, Aerts I, … Geoerger B (2022). Cobimetinib in pediatric and young adult patients with relapsed or refractory solid tumors (iMATRIX-cobi): A multicenter, phase I/II study. Targeted Oncology, 17(3), 283–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Turski ML, Brady DC, Kim HJ, Kim BE, Nose Y, Counter CM, … Thiele DJ (2012). A novel role for copper in Ras/mitogen-activated protein kinase signaling. Molecular and Cellular Biology, 32(7), 1284–1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Uko NE, Guner OF, Matesic DF, & Bowen JP (2020). Akt pathway inhibitors. Current Topics in Medicinal Chemistry, 20(10), 883–900. [DOI] [PubMed] [Google Scholar]
  112. Ursino M, & Stallard N (2021). Bayesian approaches for confirmatory trials in rare diseases: Opportunities and challenges. International Journal of Environmental Research and Public Health, 18(3), 1–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Venot Q, Blanc T, Rabia SH, Berteloot L, Ladraa S, Duong JP, … Canaud G (2018). Targeted therapy in patients with PIK3CA-related overgrowth syndrome. Nature, 558(7711), 540–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Wang D, Wang Q, Wang Y, Chen P, Lu X, Jia F, … Zhang K (2022). Targeting oncogenic KRAS with molecular brush-conjugated antisense oligonucleotides. Proceedings of the National Academy of Sciences of the United States of America, 119(29), e2113180119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Wang J, Chandrasekhar V, Abbadessa G, Yu Y, Schwartz B, & Kontaridis MI (2017). In vivo efficacy of the AKT inhibitor ARQ 092 in Noonan syndrome with multiple lentigines-associated hypertrophic cardiomyopathy. PLoS One, 12(6), e0178905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Wang RR, Liu WS, Zhou L, Ma Y, & Wang RL (2020). Probing the acting mode and advantages of RMC-4550 as an Src-homology 2 domain-containing protein tyrosine phosphatase (SHP2) inhibitor at molecular level through molecular docking and molecular dynamics. Journal of Biomolecular Structure & Dynamics, 38(5), 1525–1538. [DOI] [PubMed] [Google Scholar]
  117. Wang X, Allen S, Blake JF, Bowcut V, Briere DM, Calinisan A, … Marx MA (2022). Identification of MRTX1133, a noncovalent, potent, and selective KRAS(G12D) inhibitor. Journal of Medicinal Chemistry, 65(4), 3123–3133. [DOI] [PubMed] [Google Scholar]
  118. Weiss BD, Wolters PL, Plotkin SR, Widemann BC, Tonsgard JH, Blakeley J, … Fisher MJ (2021). NF106: A neurofibromatosis clinical trials consortium phase II trial of the MEK inhibitor mirdametinib (PD-0325901) in adolescents and adults with NF1-related plexiform neurofibromas. Journal of Clinical Oncology, 39(7), 797–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Wilkinson JD, Lowe AM, Salbert BA, Sleeper LA, Colan SD, Cox GF, … Lipshultz SE (2012). Outcomes in children with Noonan syndrome and hypertrophic cardiomyopathy: A study from the Pediatric Cardiomyopathy Registry. American Heart Journal, 164(3), 442–448. [DOI] [PubMed] [Google Scholar]
  120. Williams B, Taylor A, Orozco O, Owen C, Kelley E, Lescarbeau A, … Bergstrom DA (2022). Abstract 3327: Discovery and characterization of the potent, allosteric SHP2 inhibitor GDC-1971 for the treatment of RTK/RAS driven tumors. Cancer Research, 82, 3327. [Google Scholar]
  121. Witzig TE, Reeder C, Han JJ, LaPlant B, Stenson M, Tun HW, … Gupta M (2015). The mTORC1 inhibitor everolimus has antitumor activity in vitro and produces tumor responses in patients with relapsed T-cell lymphoma. Blood, 126(3), 328–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Wolf CM, Zenker M, Burkitt-Wright E, Edouard T, Garcia-Minaur S, Lebl J, … Ostman-Smith I (2022). Management of cardiac aspects in children with Noonan syndrome - results from a European clinical practice survey among paediatric cardiologists. European Journal of Medical Genetics, 65(1), 104372. [DOI] [PubMed] [Google Scholar]
  123. Wong JC, Perez-Mancera PA, Huang TQ, Kim J, Grego-Bessa J, Del Pilar AM, … Shannon K (2020). KrasP34R and KrasT58I mutations induce distinct RASopathy phenotypes in mice. JCI Insight, 5(21), 1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Wu X, Simpson J, Hong JH, Kim KH, Thavarajah NK, Backx PH, … Araki T (2011). MEK-ERK pathway modulation ameliorates disease phenotypes in a mouse model of Noonan syndrome associated with the Raf1(L613V) mutation. The Journal of Clinical Investigation, 121(3), 1009–1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Xu M, Casio M, Range DE, Sosa JA, & Counter CM (2018). Copper chelation as targeted therapy in a mouse model of oncogenic BRAF-driven papillary thyroid cancer. Clinical Cancer Research, 24(17), 4271–4281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Yang Q, Modi P, Newcomb T, Queva C, & Gandhi V (2015). Idelalisib: First-in-class PI3K delta inhibitor for the treatment of chronic lymphocytic leukemia, small lymphocytic leukemia, and follicular lymphoma. Clinical Cancer Research, 21(7), 1537–1542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Yen I, Shanahan F, Lee J, Hong YS, Shin SJ, Moore AR, … Malek S (2021). ARAF mutations confer resistance to the RAF inhibitor belvarafenib in melanoma. Nature, 594(7863), 418–423. [DOI] [PubMed] [Google Scholar]
  128. Yi JS, Huang Y, Kwaczala AT, Kuo IY, Ehrlich BE, Campbell SG, … Bennett AM (2016). Low-dose dasatinib rescues cardiac function in Noonan syndrome. JCI Insight, 1(20), e90220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Yi JS, Perla S, Huang Y, Mizuno K, Giordano FJ, Vinks AA, & Bennett AM (2022). Low-dose dasatinib ameliorates hypertrophic cardiomyopathy in Noonan syndrome with multiple lentigines. Cardio-vascular Drugs and Therapy, 36(4), 589–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Yu Y, Savage RE, Eathiraj S, Meade J, Wick MJ, Hall T, … Schwartz B (2015). Targeting AKT1-E17K and the PI3K/AKT pathway with an allosteric AKT inhibitor, ARQ 092. PLoS One, 10(10), e0140479. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Yuan X, Bu H, Zhou J, Yang CY, & Zhang H (2020). Recent advances of SHP2 inhibitors in cancer therapy: Current development and clinical application. Journal of Medicinal Chemistry, 63(20), 11368–11396. [DOI] [PubMed] [Google Scholar]
  132. Zhang Z, Guiley KZ, & Shokat KM (2022). Chemical acylation of an acquired serine suppresses oncogenic signaling of K-Ras(G12S). Nature Chemical Biology, 18, 1177–1183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Zhao Y, & Adjei AA (2014). The clinical development of MEK inhibitors. Nature Reviews Clinical Oncology, 11(7), 385–400. [DOI] [PubMed] [Google Scholar]
  134. Zhou P, Xiao M, Li W, Sun X, Bai Y, Meng F, … Sun K (2022). SHP2 inhibitors show anti-myeloma activity and synergize with Bortezomib in the treatment of multiple myeloma. Frontiers in Pharmacology, 13, 841308. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

This manuscript does not contain data that would require public availability.

RESOURCES