Skip to main content

Recommendations for the diagnosis and management of childhood Prader-Willi syndrome in China

Abstract

Prader-Willi syndrome (PWS) is a complex and multisystem neurobehavioral disease, which is caused by the lack of expression of paternally inherited imprinted genes on chromosome15q11.2-q13.1. The clinical manifestations of PWS vary with age. It is characterized by severe hypotonia with poor suck and feeding difficulties in the early infancy, followed by overeating in late infancy or early childhood and progressive development of morbid obesity unless the diet is externally controlled. Compared to Western PWS patients, Chinese patients have a higher ratio of deletion type. Although some rare disease networks, including PWS Cooperation Group of Rare Diseases Branch of Chinese Pediatric Society, Zhejiang Expert Group for PWS, were established recently, misdiagnosis, missed diagnosis and inappropriate intervention were usually noted in China. Therefore, there is an urgent need for an integrated multidisciplinary approach to facilitate early diagnosis and optimize management to improve quality of life, prevent complications, and prolong life expectancy. Our purpose is to evaluate the current literature and evidences on diagnosis and management of PWS in order to provide evidence-based guidelines for this disease, specially from China.

Introduction

Prader-Willi syndrome (PWS, ORPHA:739) is a complex and multisystem neurobehavioral disorder, which is caused by the lack of expression of paternally inherited imprinted genes on chromosome15q11.2-q13.1 [1, 2]. Down JL. first described the clinical manifestations of PWS in an adolescent female in 1887 [3]. However, this syndrome was not recognized until 1956, when Prader A. and Willi H. reported nine individuals with similar clinical findings [4]. Its estimated incidence ranges from 1 in 10,000 to 1 in 30,000, with equal number of males and females affected [5]. The clinical features of PWS vary with age. It is characterized by severe hypotonia with poor suck and feeding difficulties in the early infancy [6], followed by overeating in late infancy or early childhood and progressive development of morbid obesity unless the diet is externally controlled. Motor milestones and language development are delayed, and all individuals have some degree of cognitive impairment. A distinctive behavioral phenotype is common, with temper tantrums, stubbornness, and manipulative and compulsive behaviors. Hypogonadism occurs in both males and females and manifests as genital hypoplasia, underdevelopment of puberty, and, in most cases, infertility. Short stature is common and associated with growth hormone (GH) deficiency (GHD). Typical facial features, strabismus and scoliosis are common, and there is an increased incidence of sleep disorders and type 2 diabetes mellitus, especially in obese patients [7, 8]. PWS is the most common genetic cause of severe obesity in children. Recently, some rare disease networks, including PWS Cooperation Group of Rare Diseases Branch of Chinese Pediatric Society, Zhejiang Expert Group for PWS, were established. Early diagnosis and treatment can significantly improve the prognosis. However, misdiagnosis, missed diagnosis and inappropriate intervention were usually noted. Therefore, there is an urgent need for an integrated multidisciplinary approach to facilitate early diagnosis and optimize management to improve quality of life, prevent complications, and prolong life expectancy. Our purpose is to evaluate the current literature and evidences on diagnosis and management of PWS in order to provide evidence-based guidelines for this disease.

Clinical characteristics

Clinical features

The clinical features of PWS vary with age. It is characterized by severe hypotonia with poor suck and feeding difficulties in the early infancy [6, 9, 10], followed by polyphagia in late infancy or early childhood and progressive development of morbid abdominal obesity unless the diet is externally controlled. The main clinical manifestations of perinatal period are decreased fetal movement, polyhydramnios, breech presentation and non-term delivery. While in infancy, PWS is characterized by hypotonia, feeding problems with a poor sucking reflex, central sleep apnea [11], hypotonia, motor delays and temperature instability [12].

The characteristic facial features of PWS include dolichocephaly or a narrow head shape, a narrow bifrontal diameter, strabismus, almond-shaped palpebral fissure, small upturned nose, down-turned corners of the mouth with xerostonia and abnormality of the dentition, hypopigmentation of the skin comparing with other family members. Physical manifestations include hypopigmentation of the skin and hair, hypogonadism with clitoral hypoplasia and hypoplastic labia in females and a small penis and cryptorchidism in males, short stature, small hands and short feet, and motor delay [13].

In early childhood (about 2–6 years of age), additional features appear including temper tantrums, food seeking behavior and polyphagia leading to abdominal obesity, if the diet is not externally controlled. In addition, intellectual disabilities, specific learning disabilities, behavioral abnormalities including repetitions, compulsions, emotional outbursts and skin picking complicated by a high pain threshold, also develop during childhood [14,15,16]. A study of 31 Chinese patients with PWS suggested that there was a lower incidence of short stature in Chinese patients with PWS than that in other studies [17, 18], although none of the participants was treated with growth hormone, which may be one of the unique features of Chinese PWS population [19]. Additionally, it was also reported that dysmorphic face, and skin picking lesions are less common among Chinese patients with PWS than that in Western patients with PWS [19].

Genotype

Genetically, PWS is an imprinted disease owing to the lack of expression of genes inherited from the paternal chromosome 15q11.2-q13.1 region. Our understanding of the molecular basis of PWS has changed dramatically with molecular genetic studies. The most common molecular mechanism of PWS is found in about 65%-75% of cases who show a de novo typical deletion on the paternal chromosome 15q11.2-q13.1 region, including two main subtypes. Subtype Ia deletion is larger, involving chromosome 15q proximal breakpoint (BP), BPI, to a distal breakpoint, BPIII. Subtype Ib deletion is smaller, involving BPII to BP III [20]. Comparing to the typical subtype Ia or Ib deletion, some atypical cases show smaller or greater deletion in size. Very few can be classified as subtype Ic (BP I-BP IV) and subtype Id (BP I-BP V) [2, 21,22,23]. An unusual or atypical deletion is seen in about 5% of PWS individuals. A small microdeletion (about 118 Kb) within the interval between SNRPN and UBE3A that spans the SNORD116 clusters and its host transcripts is also an etiological factor in PWS [24, 25]. The second most common molecular mechanism of PWS is found in nearly 20%-30% of cases and associated with maternal uniparental disomy (mUPD) 15, in which both copies of chromosome 15 come from the mother. It includes two subtypes. Subtype IIa is isodisomy in which both chromosome 15 come from the grandmother or grandfather. While the subtype IIb is heterodisomy in which one chromosome 15 is from the grandmother and the other is from the grandfather. Due to the imprinted gene regulation, the same genes in the maternal chromosome, 15q11.2-q13.1, are intact in structure but repressed at the transcriptional level because of the epigenetic mechanism, which is primarily by methylation [26]. The third molecular mechanism of PWS is less common and is found in about 3% patients with PWS, termed imprinting defect (ID), including epimutation and the PWS imprinting center (PWS-IC) deletion. The epigenotype measured by DNA methylation for both chromosomes in the 15q11.2-q13.1 region is maternal. Some other rare genetic alterations and molecular mechanisms of PWS have been reported, such as Robertsonian translocation (15;15) inherited from the mother. Except for the pathogenic variants in the MAGEL2 gene that have been associated with Schaaf-Yang syndrome [27], no pathogenic variants of SNRPN was reported in PWS patients [28, 29] as showed in Table 1. It was reported that there was a higher incidence of paternal deletion in Chinese patients with PWS than that in Western patients with PWS and there was also a correspondingly lower incidence of mUPD [19], which were consistent with those of other Asian studies [17, 30, 31].On rare occasions, a second chromosomal anomaly may be found in addition to the 15q11.2 deletion, such as Klinefelter syndrome [32,33,34,35]. Therefore, counseling families about the clinical prognosis of the proband can be affected by the presence of additional chromosomal abnormalities.

Table 1 The genotype of Prader-Willi syndrome

Genotype–phenotype correlations

Although it was reported that there were differences in the frequency or severity of certain features among different genotypes, mainly in the paternal deletion and mUPD 15, there was no specific phenotypic feature which was known to be associated with either of the 2 main genotypes. It was reported that PWS patients with a paternal deletion had more prominent feeding problems, speech articulation impairment and sleep disturbance [36, 37]. A study of 31 Chinese patients with PWS showed that small hands and feet were more common in patients with paternal deletions than that in patients with mUPD. Besides, patients with paternal deletions were also more likely to be associated with hyperphagia and excessive weight gain than that patients with mUPD [19]. Additionally, there were differences between the paternal deletion and mUPD 15 in maladaptive behaviors. Compared with the mUPD 15, the paternal deletion type had higher self-injury and stealing scores [38]. While individuals with mUPD 15 had a slightly higher verbal IQ and milder behavior problems [39, 40]. However, it was reported that patients with mUPD 15 were at greater risk of having a psychosis in adulthood [41,42,43,44] and autism spectrum disorder (ASD) [45,46,47,48]. In one study, compared to 47% with a paternal deletion, as many as 74% individuals with mUPD 15 had been prescribed psychotropic medication [14]. However, there were also some reports of no difference between the two genotypes.

Diagnosis

The proposed clinical diagnostic criteria have changed over the past few decades [18]. A consensus was established in 1993 when genetic testing was very limited and updated in 2001 [18, 49]. The age of diagnosis has dropped significantly over the past decade, with most cases now being diagnosed in the first few months of life [50]. This allows the earlier introduction of treatments in order to reduce the morbidity particularly by preventing obesity, which will not only improve the quality of life of patients with PWS, but also reduce the burden on families and caregivers [51, 52]. The diagnosis of PWS relies on a combination of clinical features and genetic analysis [10, 13].

Consensus diagnostic criteria

Consensus clinical diagnostic criteria for PWS using a numerical scale was developed in 1993 before the availability of diagnostic testing and later revised with updates based on clinical findings and presentation in 2001 [18, 49], which included 8 major criteria. while in 2012, Cassidy SB changed it to 6 [5]. To score, major criteria are weighted at 1 point each, and minor criteria are weighted at 1⁄2 point each. Supportive findings increase the certainty of diagnosis but are not scored. It requires 5 points (at least 4 of them major) for children 3 years of age or younger and 8 points (at least 5 of them major) for children 3 years of age or older, as showed in Table 2. However, confirmation of the diagnosis of PWS requires molecular genetic testing [53].

Table 2 Consensus diagnostic criteria for Prader-Willi syndrome

Genetic testing methods

There are various methods to identify genetic alterations in PWS patients using peripheral blood lymphocytes [54, 55]. The DNA methylation pattern of the promoter-exon 1 region of the SNURF-SNRPN bicistronic gene (15q11.2) was used as the most sensitive laboratory method for the diagnosis of PWS [56,57,58]. Options for DNA methylation analysis include methylation-specific polymerase chain reaction (MS-PCR) and methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA) [59, 60]. Further approaches can be performed to identify the genetic types and allow appropriate genetic counseling, particularly for the recurrence risk. High-resolution chromosomal microarray analysis (CMA) could be used to determine the presence and size of a chromosome deletion as well as to identify partial mUPD 15 subtype [22]. Fluorescence in-situ hybridization (FISH) with high resolution karyotype may identify the deletion status or rule out a chromosomal rearrangement. However it is used less often nowadays. Although DNA sequencing may detect microdeletion, DNA sequence analysis can be considered for rare situations [61, 62], as showed in Table 3. DNA polymorphism or linkage analysis can be performed to determine whether the inheritance pattern is biparental (normal) or maternal-only (maternal disomy).

Table 3 Genetic testing used in Prader-Willi syndrome

Procedure of molecular diagnosis

DNA methylation analysis is the preferred method for the diagnosis of PWS, which detects more than 99% of the cases, including deletions, mUPD and IC defects (Fig. 1). Common options for DNA methylation analysis include MS-PCR and MS-MLPA. Compared with MS-PCR, MS-MLPA has the advantage of identifying the DNA methylation status as well as deletions. Therefore, MS-MLPA is preferred for the molecular diagnosis of PWS. Hypermethylation with copy number loss implies a paternal deletion while hypermethylation with normal copy number implies the mUPD, epimutation or Robertsonian translocation. FISH or high-resolution karyotype can be performed to distinguish translocation from mUPD and epimutation, if necessary. If the result of FISH is normal, then the DNA polymorphism or linkage analysis can be performed to distinguish mUPD from epimutation. If the result of MS-MLPA is normal, DNA sequence analysis can be performed to identify IC deletion and key genes pathogenic variants for patients highly suspected for PWS. If the result of the DNA sequence analysis is also normal, then additional tests are necessary to perform for further diagnosis.

Fig. 1
figure 1

Algorithm for genetic testing for Prader-Willi syndrome. MS-MLPA, methylation-specific-multiplex ligation probe amplification; FISH, fluorescence in situ hybridization; IC, imprinting center; SNP, single nucleotide polymorphism; mUPD, maternal uniparental disomy

MS-PCR can be performed first if MS-MLPA is not available [63]. If the result of MS-PCR is abnormal, then CMA with single nucleotide polymorphism (SNP) and copy number variant (CNV) probes can be used to determine the copy number, which may distinguish the deletion type from the other subtypes (e.g. mUPD, epimutation) [2, 59, 60, 64]. If the result of MS-PCR is normal, DNA sequence analysis may also need to identify IC deletion and key genes pathogenic variants. Furthermore, when testing for PWS does not explain all the features, additional tests are also in need to perform for further diagnosis.

After a molecular diagnosis is determined for an affected child, it is highly recommended to test the asymptomatic parents in order to determine the origin of the genetic alteration and the recurrence risk for the purpose of genetic counseling. Although majority of PWS have a low recurrence risk (< 1%), some genetic etiology could lead to high recurrence risk (> 50%), such as paternal balanced translocations or maternal 15/15 Robertsonian translocation.

Prenatal diagnosis and newborn screening

Prenatal diagnosis is not recommended routinely yet, although it can theoretically be suspected in cases of reduced fetal movement and polyhydramnios [6, 65,66,67]. Genetic testing can be performed on samples obtained from chorionic villus sampling and amniocentesis [68]. FISH or CMA can easily pick up deletions from such tissues [69], but DNA methylation analysis is necessary in cases of mUPD and ID [57]. However, clinical laboratories that perform prenatal DNA methylation analysis are reluctant to use such chorionic villus samples because of the corresponding hypomethylation of the tissue. The introduction of techniques such as comparative array genomic hybridization can be used to prenatal diagnosis of PWS owing to deletions [70].

Although it can theoretically be suspected in cases of hypotonia (for both sexes) and testicular ectopy (in boys), the clinical diagnosis of PWS in newborns is challenging because the distinctive phenotypic characteristics of the disease are not completely evident during this phase. Reliable and low-cost molecular analysis techniques are imperative for accurate and early diagnosis to start precise treatment. It was reported that DNA extracted from dried blood spot (DBS) could be analyzed by the methylation-sensitive high-resolution melting (MS-HRM) methodology, which used a unique pair of primers to amplify the promoter-exon 1 region of the SNURF-SNRPN locus and reveal its methylation status [57], providing an accurate approach for genetic screening of PWS in newborns [71]. Compared to the traditional whole blood methods, the use of the DBS sample as the main source of DNA provides several advantages: demanding only a small amount of blood, less invasive procedure, ease of storage, and transportation [72]. However, the technology is not widely available.

Differential diagnosis

Patients with negative testing for PWS should be investigated for other chromosomal deletions and duplications associated with PWS-like characteristics [27, 73], as well as possible single-gene defects [74,75,76,77].

Hypotonia in infancy is seen in many other conditions [6, 78, 79]. The common causes are neonatal sepsis and central nervous system depression. Congenital myotonic dystrophy type I is characterized by hypotonia and severe generalized weakness at birth, often accompanied by respiratory insufficiency, developmental delay, and early death [79, 80]. Additionally, a lot of muscular and neurological diseases are characterized by neonatal hypotonia, including some spinal muscular atrophy. In these cases, there may be shortness of breath, which is rarely seen in patients with PWS. Molecular genetic testing, electromyography, and/or muscle biopsy are usually required to differentiate these diseases. Many congenital metabolic errors may also manifest as hypotonia and are found to be lethargy or non-lethargy in infancy. Pompe disease is in particular worth considering [81].

Several genetic disorders are characterized by obesity, developmental delay, intellectual disability with or without hypogonadism. Angelman syndrome (AS) and Fragile X syndrome can both include obesity in a subset of individuals but no hypogonadism [82, 83]. mUPD for chromosome 14 causes early motor and speech delay, excess weight, hypotonia, and can also present with feeding problems, short stature, small hands and feet, and scoliosis [84, 85]. However, the typical clinical presentation of it is early puberty and joint laxity.

Many cytogenetic abnormalities result in overlapping manifestations with PWS, such as deletion of 1p36, 2q37.3, 6q16.2, and 10q26 and duplication of 3p25.3.26.2 and Xq27.2-ter [75, 86,87,88,89]. Therefore, when testing for PWS is negative or does not explain all the features, CMA is an appropriate test. Careful clinical evaluation by a medical geneticist or other trained diagnostician is highly recommended to direct testing appropriately and may avoid the unnecessary expense of molecular testing for diagnoses which are less likely based on clinical findings.

Management

Management of the manifestations of PWS depends on age and should include guidance to manage the consequences and expectations of the syndrome. Multidisciplinary treatment has been shown to reduce hospital stays and prevent early obesity.

Focus on different times

In infancy, in order to assure adequate nutrition, special nipples or gavage feeding is usually needed because poor suck will lead to developmental delay if they are untreated. Growth measurements including height, weight, and head circumference should be obtained and plotted at diagnosis [90]. The adrenal cortex function should also be noticed. In addition, physical therapy may improve the muscle strength. If individuals have cryptorchidism, hormonal and surgical treatment should be considered.

In childhood (after 1 year of age), guardians should strictly supervise the daily food intake to avoid excessive weight gain and encourage physical activity. Guardians are advised to take an educational plan for their children [91]. Replacement of sex hormones at puberty is beneficial to produce adequate secondary sexual characteristics, and should be supervised by a pediatric endocrinologist. Adrenal function should be reassessed later in the adolescence or adulthood (even if it is normal in infants).

In adulthood, it is also of great importance for PWS individuals to regulate behavior and manage weight to prevent morbid obesity. Involvement of occupational therapy helps with transitioning to independence in areas such as self-care and performing other activities of daily living.

GH treatment

Recombinant human GH (rhGH) therapy in patients with PWS has been used by the medical community since its approval in the United States in 2000 and in Europe in 2001 [92]. It is beneficial to normalize height, increase lean body mass, flexibility and activity levels, and reduce fat mass [93,94,95,96,97,98,99].

No consensus was reached on the age of starting rhGH treatment, although all agreed to the benefits of treating before the onset of obesity. Researches have suggested that the earlier growth hormone therapy is started, the more benefit it has (often around 3–6 month of age) [96, 100, 101]. Before initiation of rhGH therapy, patients with PWS should have a confirmed molecular diagnosis and expert multidisciplinary evaluation. GH stimulation testing should not be required as part of the therapeutic decision-making process in infants and children with PWS [102]. Selection of patients with PWS for rhGH therapy and dosing strategy should not depend on the genetic class of PWS. However, when patients with PWS have one or more of the following conditions: severe obesity, uncontrolled diabetes, untreated severe obstructive sleep apnea [11], active cancer, and active psychosis, the treatment of rhGH should be under careful consideration. Scoliosis should not be considered as a contraindication to rhGH treatment in patients with PWS.

Infants and children with PWS should start with a daily dose of 0.5 mg/m2·d with subsequent adjustments toward 1.0 mg/m2·d according to clinical response and be guided by maintenance of physiological levels of insulin like growth factor 1 (IGF-1). IGF-1 levels in patients with rhGH treatment should be maintained within the upper part of normal range (maximum + 2SDS) for healthy, age-matched normal individuals. Clinical outcome priorities should vary depending on age and on the presence of physical, mental, and social disability. Treatment with rhGH must be in the context of appropriate dietary, environmental, and lifestyle interventions necessary for care of all patients with PWS. Adults with PWS should receive a starting dose of 0.1–0.2 mg/d based on age, presence of edema, prior rhGH exposure and sensitivity, and concomitant oral estrogen use. Subsequent dosage titration should be based on clinical response, age-, and sex-appropriate IGF-1 levels in the 0 to + 2 SDS range [102, 103].

It seems that GH treatment is generally safe and well tolerated [104]. Thyroid function should be estimated and levothyroxine should be supplied before GH treatment if hypothyroidism was noted. Although GH treatment could result in an increase in fasting plasma glucose levels and insulin resistance, it usually does not increase the onset of diabetes which seems to be most influenced by obesity [103, 105]. Additionally, because there have been reports of accidental deaths in patients with PWS during GH treatment, monitoring for breathing problems and sleep apnea is recommended [106]. Other potential side effects include joint pain, edema, scoliosis and intracranial hypertension [104]. Therefore, it is recommended to perform polysomnography before starting treatment and periodically after, monitoring growth rate, glucose profile, IGF-1 levels, thyroid function, monitoring development and/or progression of scoliosis during growth [107], and liver function (especially for individuals under one year old), as showed in Table 4.

Table 4 Evaluation of patients with Prader-Willi syndrome during rhGH treatment

Hypogonadism

Unilateral or bilateral cryptorchidism is a common finding in male patients with PWS. Cryptorchidism should be sought and addressed with hormonal and/or surgical treatments. The early treatment of hypogonadism (within the first 6 months of life) is beneficial to many male patients with PWS. The treatment includes human chorionic gonadotropin (hCG) in order to improve phallus size and assist with testicular descent into the scrotal sac in few patients [93]. Surgical correction of cryptorchidism should be completed on the first or at latest in the second year of life [108].

Hypogonadism is present in both males and females and has both a primary and central (hypothalamic) etiology, with the latter believed to be more influential. Although individuals with PWS do enter puberty, the progress is arrested, and puberty is not complete. Treatment of hypogonadism usually commences at around age 11–12 years for females and age 12–13 for males, if it was desired by the parents and child. It is recommended that females can be treated with low-dose estrogen therapy with escalating doses for 2 years or until menarche, at which point they are transitioned to a combined estrogen-progesterone oral contraceptive pill or transdermal patch [5]. The decision to treat hypogonadism in females with PWS is a personal decision for each family and usually relies on the maturity level, independence, and degree of obsessive–compulsive behaviors in PWS patients. Adolescent males with PWS can be treated with either a low-dose transdermal testosterone patch or gel with escalating doses every 3–6 months to allow the testosterone levels to get into the normal range for age or with hCG therapy [109]. Oral testosterone can induce secondary sexual characteristics. GnRH or hCG therapy increases endogenous testosterone production, which can increase testicular volume and lean body mass, but not cause the mood and aggression problems that are characteristic of testosterone therapy [110, 111]. Sex education and contraception should be considered, especially for PWS women, as pregnancy has been reported infrequently [112, 113].

Precocious pubarche is not uncommon in PWS. It is considered to be secondary to obesity and premature adrenarche [114]. Premature adrenarche in PWS is not rapidly progressively or associated with other signs of central precocious puberty, and commonly develops genuinely [114]. Further investigations or treatments are usually not required. Central precocious puberty is very rare in patients with PWS [114].

Others

At present, the management of PWS is based on symptoms. It was reported that Limosilactobacillus reuteri probiotic could be used to modulate BMI, social behaviors, and gut microbiota in patients with PWS, although further investigation was warranted [115]. Oxytocin (OXT) and its analogue, carbetocin, are extensively studied in PWS clinical trials and have shown potential for treating both hyperphagia and behavior problems in pediatric PWS, although future investigations should confirm the previous study findings with extended follow-up periods within larger, well-defined clinical cohorts and also determine long-term effects and safety [116, 117]. On the basis of a phase 2 clinical trial it is demonstrated that Diazoxide Choline Controlled-Release (DCCR) decreased appetite-related behaviors and fat mass in patients with PWS [118]. Calcium and vitamin D supplements should be considered to optimize attainment of peak bone mass [108]. N-acetylcysteine or topiramate can be used to reduce skin picking [119,120,121], and recently is reported to be efficient against hyperphagia on Dykens questionnaire (severity and behavior) [122]. Additionally, it was reported that modafinil could be used to treat daytime sleepiness [123]. Although bariatric surgery is currently the most effective therapy to induce weight loss in patients with morbid obesity, its use in PWS remains controversial. It was reported that bariatric surgery could not produce sustainable long-term weight loss or comorbidity resolution in PWS and suggested that bariatric surgery could not be recommended to patients with PWS as a standard treatment [124]. MetAP2 inhibition with beloranib is reported to produce statistically significant and clinically meaningful improvements in hyperphagia-related behaviors and weight loss in participants with PWS [125]. There are also many other new therapies currently in development ranging from very early discovery and preclinical studies to active clinical trials, such as genetic therapies (gene activation by small molecules, CRISPR-based activation, oligonucleotide therapy, AAV-based gene activation, epigenome editing), hyperphagia/obesity drugs (e.g. setmelanotide, tesomet, cannabidivarin, aardvark 101, GLP receptor agonists), devices (e.g. transcranial direct current stimulation) (https://www.fpwr.org/therapeutics-in-development-for-pws).

Routine vaccination is recommended for patients with PWS when their conditions are stable and there are no other contraindications, such as severe malnutrition or infection.

Genetic counseling

All patients need genetic counseling. It is critical to know the specific genetic etiology in patients with PWS for the appropriate genetic counseling of affected families. Most families have a recurrence risk less than 1%. According to the genetic mechanism and recurrence risk, PWS was divided in 3 types, which is some different from above mentioned genotypes (Table 1). Almost all 15q11.2-q13 deletions are de novo interstitial paternal deletions (Ia), which have a very low recurrence risk (< 1%). A deletion due to chromosomal rearrangement (Ib) has a recurrence risk possibly up to 50%. Therefore, it is necessary to exclude balanced translocation for the father of deletion type PWS patient by FISH, or whole exome sequencing which is still rarely used because of the high cost.

mUPD 15 is typically de novo (IIa), with a recurrence ratio < 1% except if there is a Robertsonian translocation (IIb) mechanism. It is almost 100% recurrence ratio if mother has a 15;15 Robertsonian translocation. Therefore, it is necessary to exclude Robertsonian translocation (15;15) for the mother of PWS by high resolution karyotype or FISH. Rarely, a small marker chromosome is also present in a proband with mUPD 15 [126]. In these instances, it is of great importance to examine both parents’ karyotype because it seems that these small marker chromosomes may increase the risk for nondisjunction and UPD [127].

Patients with PWS due to an ID should be tested for an IC deletion by a laboratory which is experienced in detecting them. The majority (approximately 85%) of those with an ID have a de novo epigenetic pathogenic variant (IIIb) and the recurrence risk is < 1% for this group. However, approximately 15% of those have ID with IC deletion (IIIa). In approximately half of these individuals, the IC deletion is familial and the recurrence risk is 50% for these families, as showed in Table 5. Therefore, fathers of children with an IC deletion should have DNA methylation and dosing analysis (or sequence analysis) to determine whether they carry the IC deletion.

Table 5 Risks to sibs of a proband with Prader-Willi syndrome by genetic mechanism

With rare exception, individuals with PWS do not reproduce. However, there were two reported female patients with genetically confirmed PWS who have had a child [112, 113]. No genetically confirmed males with PWS have been known to have fathered a child. The risk of the child of an affected individual depends on the etiology of the PWS and the gender of the affected individual. The offspring have a 50% risk of having AS, if the female proband has PWS with a deletion, due to the loss of function of the maternal copy of the UBE3A which is also located on the 15q11.2-q13.1. Almost all offspring of PWS patients with Robertsonian translocation are PWS or AS due to trisomy rescue or monosomy.

Conclusions

The complex genetics, etiology, multiple phenotypes, and evolving natural history of PWS mean that a multidisciplinary professional, parental, societal, and environmental approach to the management is required with many challenges to reduce morbidity and mortality and improve quality of life. However, in recent years, there has been increasing appreciation and availability of important management strategies, which have already made significant improvements in the life of individuals with PWS. The management strategies include early diagnosis, evaluation and treatment by multidisciplinary teams, introduction of GH treatment, control of the food environment, and better understanding of the behavioral and psychiatric aspects. Whereas filling of the gaps in our understanding of the underlying science will translate and eventually guide clinical management of PWS (e.g. identification of genes and their link to particular phenotypes, genotype–phenotype correlations), several clinical and pathophysiological questions need to be urgently addressed to continue improvement in the care of patients with PWS.

Availability of data and materials

Data sharing not applicable to this article as no datasets were generated or analysed during the current study.

References

  1. Cassidy SB, Dykens E, Williams CA. Prader-Willi and Angelman syndromes: sister imprinted disorders. Am J Med Genet. 2000;97(2):136–46.

    Article  CAS  PubMed  Google Scholar 

  2. Butler MG. Prader-Willi syndrome: obesity due to genomic imprinting. Curr Genomics. 2011;12(3):204–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Down JL. Lettsomian lectures on some of the mental affections of childhood and youth. Br Med J. 1887;1(1362):256–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ledbetter DH, Riccardi VM, Airhart SD, Strobel RJ, Keenan BS, Crawford JD. Deletions of chromosome 15 as a cause of the Prader-Willi syndrome. N Engl J Med. 1981;304(6):325–9.

    Article  CAS  PubMed  Google Scholar 

  5. Cassidy SB, Schwartz S, Miller JL, Driscoll DJ. Prader-Willi syndrome. Geneti Med. 2012;14(1):10–26.

    Article  CAS  Google Scholar 

  6. Yang L, Zhou Q, Ma B, Mao S, Dai Y, Zhu M, et al. Perinatal features of Prader-Willi syndrome: a Chinese cohort of 134 patients. Orphanet J Rare Dis. 2020;15(1):24.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Emerick JE, Vogt KS. Endocrine manifestations and management of Prader-Willi syndrome. Int J Pediatr Endocrinol. 2013;2013(1):14.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Yang-Li D, Ke H, Chao-Chun Z, Guan-Ping D. Chinese siblings with Prader-Willi syndrome inherited from their paternal grandmother. Indian Pediatr. 2019;56(9):789–91.

    Article  PubMed  Google Scholar 

  9. Zhou Y, Ma MS, Li GY, Zhang ZJ, Ding J, Xu YW, et al. Analysis of the clinical perinatal characteristics of 226 patients with Prader-Willi syndrome in China. Zhonghua er ke za zhi = Chin J Pediatr. 2021;59(6):466–70.

    CAS  Google Scholar 

  10. Dong GQ, Su YY, Qiu XY, Lu XY, Li JX, Huang M, et al. Clinical screening and genetic diagnosis for Prader-Willi syndrome. Zhongguo dang dai er ke za zhi = Chin J Contemp Pediatr. 2020;22(9):1001–6.

    Google Scholar 

  11. Lu A, Luo F, Sun C, Zhang X, Wang L, Lu W. Sleep-disordered breathing and genetic findings in children with Prader-Willi syndrome in China. Ann Transl Med. 2020;8(16):989.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Butler MG, Miller JL, Forster JL. Prader-Willi syndrome—clinical genetics, diagnosis and treatment approaches: an update. Curr Pediatr Rev. 2019;15(4):207–44.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Writing Group For Practice Guidelines For D, Treatment Of Genetic Diseases Medical Genetics Branch Of Chinese Medical A, Li C, Xie B, Shen Y, Luo F. Clinical practice guidelines for Prader-Willi syndrome. Zhonghua yi xue yi chuan xue za zhi = Zhonghua yixue yichuanxue zazhi = Chin J Med Genet. 2020;37(3):318–23.

    Google Scholar 

  14. Soni S, Whittington J, Holland AJ, Webb T, Maina E, Boer H, et al. The course and outcome of psychiatric illness in people with Prader-Willi syndrome: implications for management and treatment. J Intellect Disabil Res JIDR. 2007;51(Pt 1):32–42.

    Article  CAS  PubMed  Google Scholar 

  15. Soni S, Whittington J, Holland AJ, Webb T, Maina EN, Boer H, et al. The phenomenology and diagnosis of psychiatric illness in people with Prader-Willi syndrome. Psychol Med. 2008;38(10):1505–14.

    Article  CAS  PubMed  Google Scholar 

  16. Aman LCS, Manning KE, Whittington JE, Holland AJ. Mechanistic insights into the genetics of affective psychosis from Prader-Willi syndrome. Lancet Psychiatry. 2018;5(4):370–8.

    Article  PubMed  Google Scholar 

  17. Lin HY, Lin SP, Chuang CK, Chen MR, Yen JL, Lee YJ, et al. Genotype and phenotype in patients with Prader-Willi syndrome in Taiwan. Acta Paediatr (Oslo, Norway: 1992). 2007;96(6):902–5.

    Article  Google Scholar 

  18. Gunay-Aygun M, Schwartz S, Heeger S, O’Riordan MA, Cassidy SB. The changing purpose of Prader-Willi syndrome clinical diagnostic criteria and proposed revised criteria. Pediatrics. 2001;108(5):E92.

    Article  CAS  PubMed  Google Scholar 

  19. Lu W, Qi Y, Cui B, Chen XL, Wu BB, Chen C, et al. Clinical and genetic features of Prader-Willi syndrome in China. Eur J Pediatr. 2014;173(1):81–6.

    Article  CAS  PubMed  Google Scholar 

  20. Cheon CK. Genetics of Prader-Willi syndrome and Prader-Will-Like syndrome. Ann Pediatr Endocrinol Metab. 2016;21(3):126–35.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Angulo MA, Butler MG, Cataletto ME. Prader-Willi syndrome: a review of clinical, genetic, and endocrine findings. J Endocrinol Investig. 2015;38(12):1249–63.

    Article  CAS  Google Scholar 

  22. Butler MG, Hartin SN, Hossain WA, Manzardo AM, Kimonis V, Dykens E, et al. Molecular genetic classification in Prader-Willi syndrome: a multisite cohort study. J Med Genet. 2019;56(3):149–53.

    Article  CAS  PubMed  Google Scholar 

  23. Kim SJ, Miller JL, Kuipers PJ, German JR, Beaudet AL, Sahoo T, et al. Unique and atypical deletions in Prader-Willi syndrome reveal distinct phenotypes. Eur J Hum Genet EJHG. 2012;20(3):283–90.

    Article  CAS  PubMed  Google Scholar 

  24. Kim Y, Wang SE, Jiang YH. Epigenetic therapy of Prader-Willi syndrome. Transl Res J Lab Clin Med. 2019;208:105–18.

    CAS  Google Scholar 

  25. Bieth E, Eddiry S, Gaston V, Lorenzini F, Buffet A, Conte Auriol F, et al. Highly restricted deletion of the SNORD116 region is implicated in Prader-Willi Syndrome. Eur J Hum Genet EJHG. 2015;23(2):252–5.

    Article  CAS  PubMed  Google Scholar 

  26. Mian-Ling Z, Yun-Qi C, Chao-Chun Z. Prader-Willi syndrome: molecular mechanism and epigenetic therapy. Curr Gene Ther. 2020;20(1):36–43.

    Article  PubMed  CAS  Google Scholar 

  27. Chen X, Ma X, Zou C. Phenotypic spectrum and genetic analysis in the fatal cases of Schaaf-Yang syndrome: two case reports and literature review. Medicine. 2020;99(29):e20574.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Negishi Y, Ieda D, Hori I, Nozaki Y, Yamagata T, Komaki H, et al. Schaaf-Yang syndrome shows a Prader-Willi syndrome-like phenotype during infancy. Orphanet J Rare Dis. 2019;14(1):277.

    Article  PubMed  PubMed Central  Google Scholar 

  29. McCarthy J, Lupo PJ, Kovar E, Rech M, Bostwick B, Scott D, et al. Schaaf-Yang syndrome overview: report of 78 individuals. Am J Med Genet A. 2018;176(12):2564–74.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Ehara H, Ohno K, Takeshita K. Frequency of the Prader-Willi syndrome in the San-in district, Japan. Brain Dev. 1995;17(5):324–6.

    Article  CAS  PubMed  Google Scholar 

  31. Kim HJ, Cho HJ, Jin DK, Kwon EK, Ki CS, Kim JW, et al. Genetic basis of Prader-Willi syndrome in Korea: less uniparental disomy than has been recognized? Clin Genet. 2004;66(4):368–72.

    Article  PubMed  Google Scholar 

  32. Butler MG, Hedges LK, Rogan PK, Seip JR, Cassidy SB, Moeschler JB. Klinefelter and trisomy X syndromes in patients with Prader-Willi syndrome and uniparental maternal disomy of chromosome 15–a coincidence? Am J Med Genet. 1997;72(1):111–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Rego A, Coll MD, Regal M, Guitart M, Escudero T, García-Mayor RV. A case with 47, XXY, del(15)(q11;q13) karyotype associated with Prader-Willi phenotype. Horm Res. 1997;48(1):44–6.

    Article  CAS  PubMed  Google Scholar 

  34. Verhoeven WM, de Vries BB, Duffels SJ, Egger JI, Noordam C, Tuinier S. Klinefelter’s syndrome and Prader-Willi syndrome: a rare combination. Psychopathology. 2007;40(5):356–60.

    Article  CAS  PubMed  Google Scholar 

  35. Zou XY, Chao YQ, Zeng LH, Zou CC. Prader-Willi syndrome coincident with DiGeorge syndrome. Indian J Pediatr. 2020;87(6):471–2.

    Article  PubMed  Google Scholar 

  36. Torrado M, Araoz V, Baialardo E, Abraldes K, Mazza C, Krochik G, et al. Clinical-etiologic correlation in children with Prader-Willi syndrome (PWS): an interdisciplinary study. Am J Med Genet Part A. 2007;143A(5):460–8.

    Article  PubMed  Google Scholar 

  37. Holsen LM, Zarcone JR, Chambers R, Butler MG, Bittel DC, Brooks WM, et al. Genetic subtype differences in neural circuitry of food motivation in Prader-Willi syndrome. Int J Obes (2005). 2009;33(2):273–83.

    Article  CAS  Google Scholar 

  38. Hartley SL, Maclean WE Jr, Butler MG, Zarcone J, Thompson T. Maladaptive behaviors and risk factors among the genetic subtypes of Prader-Willi syndrome. Am J Med Genet A. 2005;136(2):140–5.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Dykens EM, Cassidy SB, King BH. Maladaptive behavior differences in Prader-Willi syndrome due to paternal deletion versus maternal uniparental disomy. Am J Ment Retard AJMR. 1999;104(1):67–77.

    Article  CAS  PubMed  Google Scholar 

  40. Copet P, Jauregi J, Laurier V, Ehlinger V, Arnaud C, Cobo AM, et al. Cognitive profile in a large French cohort of adults with Prader-Willi syndrome: differences between genotypes. J Intellect Disabil Res JIDR. 2010;54(3):204–15.

    Article  CAS  PubMed  Google Scholar 

  41. Holland AJ, Whittington JE, Butler J, Webb T, Boer H, Clarke D. Behavioural phenotypes associated with specific genetic disorders: evidence from a population-based study of people with Prader-Willi syndrome. Psychol Med. 2003;33(1):141–53.

    Article  CAS  PubMed  Google Scholar 

  42. Whittington J, Holland A. A review of psychiatric conceptions of mental and behavioural disorders in Prader-Willi syndrome. Neurosci Biobehav Rev. 2018;95:396–405.

    Article  PubMed  Google Scholar 

  43. Yang L, Zhan GD, Ding JJ, Wang HJ, Ma D, Huang GY, et al. Psychiatric illness and intellectual disability in the Prader-Willi syndrome with different molecular defects—a meta analysis. PLoS ONE. 2013;8(8):e72640.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sinnema M, Boer H, Collin P, Maaskant MA, van Roozendaal KE, Schrander-Stumpel CT, et al. Psychiatric illness in a cohort of adults with Prader-Willi syndrome. Res Dev Disabil. 2011;32(5):1729–35.

    Article  PubMed  Google Scholar 

  45. Veltman MW, Craig EE, Bolton PF. Autism spectrum disorders in Prader-Willi and Angelman syndromes: a systematic review. Psychiatr Genet. 2005;15(4):243–54.

    Article  PubMed  Google Scholar 

  46. Bennett JA, Germani T, Haqq AM, Zwaigenbaum L. Autism spectrum disorder in Prader-Willi syndrome: a systematic review. Am J Med Genet Part A. 2015;167A(12):2936–44.

    Article  PubMed  Google Scholar 

  47. Veltman MW, Thompson RJ, Roberts SE, Thomas NS, Whittington J, Bolton PF. Prader-Willi syndrome—a study comparing deletion and uniparental disomy cases with reference to autism spectrum disorders. Eur Child Adolesc Psychiatry. 2004;13(1):42–50.

    Article  PubMed  Google Scholar 

  48. Milner KM, Craig EE, Thompson RJ, Veltman MW, Thomas NS, Roberts S, et al. Prader-Willi syndrome: intellectual abilities and behavioural features by genetic subtype. J Child Psychol Psychiatry. 2005;46(10):1089–96.

    Article  PubMed  Google Scholar 

  49. Holm VA, Cassidy SB, Butler MG, Hanchett JM, Greenswag LR, Whitman BY, et al. Prader-Willi syndrome: consensus diagnostic criteria. Pediatrics. 1993;91(2):398–402.

    Article  CAS  PubMed  Google Scholar 

  50. Bacheré N, Diene G, Delagnes V, Molinas C, Moulin P, Tauber M. Early diagnosis and multidisciplinary care reduce the hospitalization time and duration of tube feeding and prevent early obesity in PWS infants. Horm Res. 2008;69(1):45–52.

    PubMed  Google Scholar 

  51. El-Bassyouni HT, Hassan N, Mahfouz I, Abd-Elnaby AE, Mostafa MI, Tosson AMS. Early detection and management of Prader-Willi syndrome in Egyptian patients. J Pediatr Genet. 2019;8(4):179–86.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Mao SJ, Shen J, Xu F, Zou CC. Quality of life in caregivers of young children with Prader-Willi syndrome. World J Pediatr. 2019;15(5):506–10.

    Article  CAS  PubMed  Google Scholar 

  53. Huang W, Li S, Luo H, Wen X, Lin C, Chen S, et al. Application of various genetic techniques for the diagnosis of Prader-Willi syndrome. Zhonghua yi xue yi chuan xue za zhi = Zhonghua yixue yichuanxue zazhi = Chin J Med Genet. 2020;37(8):875–8.

    Google Scholar 

  54. Monaghan KG, Van Dyke DL, Feldman G, Wiktor A, Weiss L. Diagnostic testing: a cost analysis for Prader-Willi and Angelman syndromes. Am J Hum Genet. 1997;60(1):244–7.

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Dupont JM, Cuisset L. Genetic basis of Prader-Willi and Angelman syndromes: implications for the biologic diagnosis. Arch Pediatr. 1998;5(4):418–24.

    Article  CAS  PubMed  Google Scholar 

  56. Dos Santos JF, Mota LR, Rocha PH, Ferreira de Lima RL. A modified MS-PCR approach to diagnose patients with Prader-Willi and Angelman syndrome. Mol Biol Rep. 2016;43(11):1221–5.

    Article  PubMed  CAS  Google Scholar 

  57. Ribeiro Ferreira I, Darleans Dos Santos Cunha W, Henrique Ferreira Gomes L, Azevedo Cintra H, Lopes Cabral Guimarães Fonseca L, Ferreira Bastos E, et al. A rapid and accurate methylation-sensitive high-resolution melting analysis assay for the diagnosis of Prader Willi and Angelman patients. Mol Genet Genom Med. 2019;7(6):e637.

    Article  CAS  Google Scholar 

  58. Muralidhar B, Butler MG. Methylation PCR analysis of Prader-Willi syndrome, Angelman syndrome, and control subjects. Am J Med Genet. 1998;80(3):263–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Bittel DC, Kibiryeva N, Butler MG. Methylation-specific multiplex ligation-dependent probe amplification analysis of subjects with chromosome 15 abnormalities. Genet Test. 2007;11(4):467–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Henkhaus RS, Kim SJ, Kimonis VE, Gold JA, Dykens EM, Driscoll DJ, et al. Methylation-specific multiplex ligation-dependent probe amplification and identification of deletion genetic subtypes in Prader-Willi syndrome. Genet Test Mol Biomark. 2012;16(3):178–86.

    Article  CAS  Google Scholar 

  61. Newkirk HL, Bittel DC, Butler MG. Analysis of the Prader-Willi syndrome chromosome region using quantitative microsphere hybridization (QMH) array. Am J Med Genet Part A. 2008;146A(18):2346–54.

    Article  CAS  PubMed  Google Scholar 

  62. Butler MG. Molecular diagnosis of Prader-Willi syndrome: comparison of cytogenetic and molecular genetic data including parent of origin dependent methylation DNA patterns. Am J Med Genet. 1996;61(2):188–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Song M, Li L, Fu J, Li X, Lu G. Detection of Prader-Willi syndrome by methylation-specific PCR. Zhonghua yi xue yi chuan xue za zhi = Zhonghua yixue yichuanxue zazhi = Chin J Med Genet. 2000;17(1):54–6.

    CAS  Google Scholar 

  64. Hartin SN, Hossain WA, Francis D, Godler DE, Barkataki S, Butler MG. Analysis of the Prader-Willi syndrome imprinting center using droplet digital PCR and next-generation whole-exome sequencing. Mol Genet Genom Med. 2019;7(4):e00575.

    Article  CAS  Google Scholar 

  65. Whittington JE, Butler JV, Holland AJ. Pre-, peri- and postnatal complications in Prader-Willi syndrome in a UK sample. Early Human Dev. 2008;84(5):331–6.

    Article  CAS  Google Scholar 

  66. Dudley O, Muscatelli F. Clinical evidence of intrauterine disturbance in Prader-Willi syndrome, a genetically imprinted neurodevelopmental disorder. Early Hum Dev. 2007;83(7):471–8.

    Article  PubMed  Google Scholar 

  67. Chen CP, Lin MH, Chen YY, Chern SR, Wu PS, Chen SW, et al. Prenatal diagnosis of a 15q11.2–q14 deletion of paternal origin associated with increased nuchal translucency, mosaicism for de novo multiple unbalanced translocations involving 15q11-q14, 5qter, 15qter, 17pter and 3qter and Prader-Willi syndrome. Taiwan J Obstet Gynecol. 2021;60(2):335–40.

    Article  PubMed  Google Scholar 

  68. Glenn CC, Deng G, Michaelis RC, Tarleton J, Phelan MC, Surh L, et al. DNA methylation analysis with respect to prenatal diagnosis of the Angelman and Prader-Willi syndromes and imprinting. Prenat Diagn. 2000;20(4):300–6.

    Article  CAS  PubMed  Google Scholar 

  69. Santa María L, Curotto B, Cortés F, Rojas C, Alliende MA. Molecular diagnosis of Prader-Willi and Angelman syndromes: methylation, cytogenetics and FISH analysis. Rev Med Chil. 2001;129(4):367–74.

    Article  PubMed  Google Scholar 

  70. Butler MG, Fischer W, Kibiryeva N, Bittel DC. Array comparative genomic hybridization (aCGH) analysis in Prader-Willi syndrome. Am J Med Genet Part A. 2008;146A(7):854–60.

    Article  CAS  PubMed  Google Scholar 

  71. Ferreira IR, Costa RA, Gomes LHF, Dos Santos Cunha WD, Tyszler LS, Freitas S, et al. A newborn screening pilot study using methylation-sensitive high resolution melting on dried blood spots to detect Prader-Willi and Angelman syndromes. Sci Rep. 2020;10(1):13026.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Mahmoud R, Singh P, Weiss L, Lakatos A, Oakes M, Hossain W, et al. Newborn screening for Prader-Willi syndrome is feasible: early diagnosis for better outcomes. Am J Med Genet A. 2019;179(1):29–36.

    CAS  PubMed  Google Scholar 

  73. Xu F, Cheng DH, Di YF, Tan K, Li LY, Lu GX, et al. Identification of a cryptic 1p36.3 microdeletion in a patient with Prader-Willi-like syndrome features. Zhonghua yi xue yi chuan xue za zhi = Zhonghua yixue yichuanxue zazhi = Chin J Med Genet. 2010;27(5):524–9.

    CAS  Google Scholar 

  74. Gilhuis HJ, van Ravenswaaij CM, Hamel BJ, Gabreëls FJ. Interstitial 6q deletion with a Prader-Willi-like phenotype: a new case and review of the literature. Eur J Paediatr Neurol EJPN. 2000;4(1):39–43.

    Article  CAS  PubMed  Google Scholar 

  75. Varela MC, Simões-Sato AY, Kim CA, Bertola DR, De Castro CI, Koiffmann CP. A new case of interstitial 6q16.2 deletion in a patient with Prader-Willi-like phenotype and investigation of SIM1 gene deletion in 87 patients with syndromic obesity. Eur J Med Genet. 2006;49(4):298–305.

    Article  PubMed  Google Scholar 

  76. Geets E, Zegers D, Beckers S, Verrijken A, Massa G, Van Hoorenbeeck K, et al. Copy number variation (CNV) analysis and mutation analysis of the 6q14.1–6q16.3 genes SIM1 and MRAP2 in Prader Willi like patients. Mol Genet Metab. 2016;117(3):383–8.

    Article  CAS  PubMed  Google Scholar 

  77. Geets E, Aerts E, Verrijken A, Van Hoorenbeeck K, Verhulst S, Van Gaal L, et al. DNA sequencing and copy number variation analysis of MCHR2 in a cohort of Prader Willi like (PWL) patients. Obes Res Clin Pract. 2018;12(2):158–66.

    Article  PubMed  Google Scholar 

  78. Miller SP, Riley P, Shevell MI. The neonatal presentation of Prader-Willi syndrome revisited. J Pediatr. 1999;134(2):226–8.

    Article  CAS  PubMed  Google Scholar 

  79. Richer LP, Shevell MI, Miller SP. Diagnostic profile of neonatal hypotonia: an 11-year study. Pediatr Neurol. 2001;25(1):32–7.

    Article  CAS  PubMed  Google Scholar 

  80. Nguyen CE, Campbell C. Myotonic dystrophy type 1. CMAJ Can Med Assoc J = journal de l’Association medicale canadienne. 2016;188(14):1033.

    Article  Google Scholar 

  81. Kohler L, Puertollano R, Raben N. Pompe disease: from basic science to therapy. Neurotherapeutics. 2018;15(4):928–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Buiting K, Williams C, Horsthemke B. Angelman syndrome - insights into a rare neurogenetic disorder. Nat Rev Neurol. 2016;12(10):584–93.

    Article  CAS  PubMed  Google Scholar 

  83. Salcedo-Arellano MJ, Hagerman RJ, Martínez-Cerdeño V. Fragile X syndrome: clinical presentation, pathology and treatment. Gac Med Mex. 2020;156(1):60–6.

    PubMed  Google Scholar 

  84. Hosoki K, Kagami M, Tanaka T, Kubota M, Kurosawa K, Kato M, et al. Maternal uniparental disomy 14 syndrome demonstrates prader-willi syndrome-like phenotype. J Pediatr. 2009;155(6):900-3.e1.

    Article  CAS  PubMed  Google Scholar 

  85. Cox H, Bullman H, Temple IK. Maternal UPD(14) in the patient with a normal karyotype: clinical report and a systematic search for cases in samples sent for testing for Prader-Willi syndrome. Am J Med Genet Part A. 2004;127A(1):21–5.

    Article  PubMed  Google Scholar 

  86. Greco M, Ferrara P, Farello G, Striano P, Verrotti A. Electroclinical features of epilepsy associated with 1p36 deletion syndrome: a review. Epilepsy Res. 2018;139:92–101.

    Article  CAS  PubMed  Google Scholar 

  87. Jordan VK, Zaveri HP, Scott DA. 1p36 deletion syndrome: an update. Appl Clin Genet. 2015;8:189–200.

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Mehraein Y, Pfob M, Steinlein O, Aichinger E, Eggert M, Bubendorff V, et al. 2q37.3 deletion syndrome: two cases with highly distinctive facial phenotype, discordant association with schizophrenic psychosis, and shared deletion breakpoint region on 2q37.3. Cytogenet Genome Res. 2015;146(1):33–8.

    Article  PubMed  Google Scholar 

  89. Beguier F, Housset M, Roubeix C, Augustin S, Zagar Y, Nous C, et al. The 10q26 risk haplotype of age-related macular degeneration aggravates subretinal inflammation by impairing monocyte elimination. Immunity. 2020;53(2):429-41.e8.

    Article  CAS  PubMed  Google Scholar 

  90. Mongkollarp N, Tim-Aroon T, Okascharoen C, Wichajarn K, Phosuwattanakul J, Chongviriyaphan N, et al. Growth charts for Thai children with Prader-Willi syndrome aged 0–18 years. Orphanet J Rare Dis. 2020;15(1):111.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Chen CM, Chen CL, Hou JW, Hsu HC, Chung CY, Chou SW, et al. Developmental profiles and mentality in preschool children with Prader-Willi syndrome: a preliminary study. Chang Gung Med J. 2010;33(4):436–42.

    PubMed  Google Scholar 

  92. Burman P, Ritzén EM, Lindgren AC. Endocrine dysfunction in Prader-Willi syndrome: a review with special reference to GH. Endocr Rev. 2001;22(6):787–99.

    Article  CAS  PubMed  Google Scholar 

  93. McCandless SE. Clinical report—health supervision for children with Prader-Willi syndrome. Pediatrics. 2011;127(1):195–204.

    Article  PubMed  Google Scholar 

  94. Longhi S, Grugni G, Gatti D, Spinozzi E, Sartorio A, Adami S, et al. Adults with Prader-Willi syndrome have weaker bones: effect of treatment with GH and sex steroids. Calcif Tissue Int. 2015;96(2):160–6.

    Article  CAS  PubMed  Google Scholar 

  95. Yang A, Choi JH, Sohn YB, Eom Y, Lee J, Yoo HW, et al. Effects of recombinant human growth hormone treatment on growth, body composition, and safety in infants or toddlers with Prader-Willi syndrome: a randomized, active-controlled trial. Orphanet J Rare Dis. 2019;14(1):216.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Corripio R, Tubau C, Calvo L, Brun C, Capdevila N, Larramona H, et al. Safety and effectiveness of growth hormone therapy in infants with Prader-Willi syndrome younger than 2 years: a prospective study. J Pediatr Endocrinol Metab JPEM. 2019;32(8):879–84.

    Article  CAS  PubMed  Google Scholar 

  97. Wolfgram PM, Carrel AL, Allen DB. Long-term effects of recombinant human growth hormone therapy in children with Prader-Willi syndrome. Curr Opin Pediatr. 2013;25(4):509–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. de Lind van Wijngaarden RF, Siemensma EP, Festen DA, Otten BJ, van Mil EG, Rotteveel J, et al. Efficacy and safety of long-term continuous growth hormone treatment in children with Prader-Willi syndrome. J Clin Endocrinol Metab. 2009;94(11):4205–15.

    Article  PubMed  CAS  Google Scholar 

  99. Yang H, Zhang M, Song H, Zhu H, Pan H. Growth patterns of Chinese patients with Prader-Willi syndrome. Congenit Anom (Kyoto). 2015;55(4):173–7.

    Article  Google Scholar 

  100. Festen DA, Wevers M, Lindgren AC, Bohm B, Otten BJ, Wit JM, et al. Mental and motor development before and during growth hormone treatment in infants and toddlers with Prader-Willi syndrome. Clin Endocrinol. 2008;68(6):919–25.

    Article  CAS  Google Scholar 

  101. Carrel AL, Myers SE, Whitman BY, Eickhoff J, Allen DB. Long-term growth hormone therapy changes the natural history of body composition and motor function in children with Prader-Willi syndrome. J Clin Endocrinol Metab. 2010;95(3):1131–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Deal CL, Tony M, Höybye C, Allen DB, Tauber M, Christiansen JS. GrowthHormone Research Society workshop summary: consensus guidelines for recombinant human growth hormone therapy in Prader-Willi syndrome. J Clin Endocrinol Metab. 2013;98(6):E1072–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Höybye C, Thorén M, Böhm B. Cognitive, emotional, physical and social effects of growth hormone treatment in adults with Prader-Willi syndrome. J Intellect Disabil Res JIDR. 2005;49(Pt 4):245–52.

    Article  PubMed  Google Scholar 

  104. Heksch R, Kamboj M, Anglin K, Obrynba K. Review of Prader-Willi syndrome: the endocrine approach. Transl Pediatr. 2017;6(4):274–85.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Bakker NE, Kuppens RJ, Siemensma EP, Tummers-de Lind van Wijngaarden RF, Festen DA, Bindels-de Heus GC, et al. Eight years of growth hormone treatment in children with Prader-Willi syndrome: maintaining the positive effects. J Clin Endocrinolo Metab. 2013;98(10):4013–22.

    Article  CAS  Google Scholar 

  106. Van Vliet G, Deal CL, Crock PA, Robitaille Y, Oligny LL. Sudden death in growth hormone-treated children with Prader-Willi syndrome. J Pediatr. 2004;144(1):129–31.

    Article  PubMed  Google Scholar 

  107. Góralska M, Bednarczuk T, Rosłon M, Libura M, Szalecki M, Hilczer M, et al. Management of Prader-Willi Syndrome (PWS) in adults—what an endocrinologist needs to know. Recommendations of the Polish Society of Endocrinology and the Polish Society of Paediatric Endocrinology and Diabetology. Endokrynol Pol. 2018. https://doi.org/10.5603/EP.2018.0047.

    Article  PubMed  Google Scholar 

  108. Goldstone AP, Holland AJ, Hauffa BP, Hokken-Koelega AC, Tauber M. Recommendations for the diagnosis and management of Prader-Willi syndrome. J Clin Endocrinol Metab. 2008;93(11):4183–97.

    Article  CAS  PubMed  Google Scholar 

  109. Eiholzer U, l’Allemand D, Rousson V, Schlumpf M, Gasser T, Girard J, et al. Hypothalamic and gonadal components of hypogonadism in boys with Prader-Labhart-Willi syndrome. J Clin Endocrinol Metab. 2006;91(3):892–8.

    Article  CAS  PubMed  Google Scholar 

  110. Eiholzer U, Grieser J, Schlumpf M, l’Allemand D. Clinical effects of treatment for hypogonadism in male adolescents with Prader-Labhart-Willi syndrome. Hormone Res. 2007;68(4):178–84.

    Article  CAS  PubMed  Google Scholar 

  111. Sinnema M, Maaskant MA, van Schrojenstein Lantman-de Valk HM, van Nieuwpoort IC, Drent ML, Curfs LM, et al. Physical health problems in adults with Prader-Willi syndrome. Am J Med Genet A. 2011;155A(9):2112–24.

    Article  PubMed  Google Scholar 

  112. Akefeldt A, Törnhage CJ, Gillberg C. A woman with Prader-Willi syndrome gives birth to a healthy baby girl. Dev Med Child Neurol. 1999;41(11):789–90.

    Article  CAS  PubMed  Google Scholar 

  113. Schulze A, Mogensen H, Hamborg-Petersen B, Graem N, Ostergaard JR, Brøndum-Nielsen K. Fertility in Prader-Willi syndrome: a case report with Angelman syndrome in the offspring. Acta Paediatr (Oslo, Norway: 1992). 2001;90(4):455–9.

    Article  CAS  Google Scholar 

  114. Crino A, Schiaffini R, Ciampalini P, Spera S, Beccaria L, Benzi F, et al. Hypogonadism and pubertal development in Prader-Willi syndrome. Eur J Pediatr. 2003;162(5):327–33.

    Article  CAS  PubMed  Google Scholar 

  115. Kong XJ, Liu K, Zhuang P, Tian R, Liu S, Clairmont C, et al. The effects of Limosilactobacillus reuteri LR-99 supplementation on body mass index, social communication, fine motor function, and gut microbiome composition in individuals with Prader-Willi syndrome: a randomized double-blinded placebo-controlled trial. Probiotics Antimicrob Proteins. 2021. https://doi.org/10.1007/s12602-021-09800-9.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Hollander E, Levine KG, Ferretti CJ, Freeman K, Doernberg E, Desilva N, et al. Intranasal oxytocin versus placebo for hyperphagia and repetitive behaviors in children with Prader-Willi Syndrome: a randomized controlled pilot trial. J Psychiatr Res. 2021;137:643–51.

    Article  PubMed  Google Scholar 

  117. Damen L, Grootjen LN, Juriaans AF, Donze SH, Huisman TM, Visser JA, et al. Oxytocin in young children with Prader-Willi syndrome: results of a randomized, double-blind, placebo-controlled, crossover trial investigating 3 months of oxytocin. Clin Endocrinol. 2021;94(5):774–85.

    Article  CAS  Google Scholar 

  118. Bischof JM, Wevrick R. Chronic diazoxide treatment decreases fat mass and improves endurance capacity in an obese mouse model of Prader-Willi syndrome. Mol Genet Metab. 2018;123(4):511–7.

    Article  CAS  PubMed  Google Scholar 

  119. Hoffman KL. Animal models of obsessive compulsive disorder: recent findings and future directions. Expert Opin Drug Discov. 2011;6(7):725–37.

    Article  CAS  PubMed  Google Scholar 

  120. Ramirez-Nino AM, D’Souza MS, Markou A. N-acetylcysteine decreased nicotine self-administration and cue-induced reinstatement of nicotine seeking in rats: comparison with the effects of N-acetylcysteine on food responding and food seeking. Psychopharmacology. 2013;225(2):473–82.

    Article  CAS  PubMed  Google Scholar 

  121. Miller JL, Angulo M. An open-label pilot study of N-acetylcysteine for skin-picking in Prader-Willi syndrome. Am J Med Genet Part A. 2014;164A(2):421–4.

    Article  PubMed  CAS  Google Scholar 

  122. Consoli A, Cabal Berthoumieu S, Raffin M, Thuilleaux D, Poitou C, Coupaye M, et al. Effect of topiramate on eating behaviours in Prader-Willi syndrome: TOPRADER double-blind randomised placebo-controlled study. Transl Psychiatry. 2019;9(1):274.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. De Cock VC, Diene G, Molinas C, Masson VD, Kieffer I, Mimoun E, et al. Efficacy of modafinil on excessive daytime sleepiness in Prader-Willi syndrome. Am J Med Genet Part A. 2011;155A(7):1552–7.

    Article  PubMed  CAS  Google Scholar 

  124. Liu SY, Wong SK, Lam CC, Ng EK. Bariatric surgery for Prader-Willi syndrome was ineffective in producing sustainable weight loss: long term results for up to 10 years. Pediatr Obes. 2020;15(1):e12575.

    Article  PubMed  Google Scholar 

  125. McCandless SE, Yanovski JA, Miller J, Fu C, Bird LM, Salehi P, et al. Effects of MetAP2 inhibition on hyperphagia and body weight in Prader-Willi syndrome: a randomized, double-blind, placebo-controlled trial. Diabetes Obes Metab. 2017;19(12):1751–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Liehr T, Brude E, Gillessen-Kaesbach G, König R, Mrasek K, von Eggeling F, et al. Prader-Willi syndrome with a karyotype 47, XY,+min(15)(pter->q11.1:) and maternal UPD 15—case report plus review of similar cases. Eur J Med Genet. 2005;48(2):175–81.

    Article  PubMed  Google Scholar 

  127. Kotzot D. Review and meta-analysis of systematic searches for uniparental disomy (UPD) other than UPD 15. Am J Med Genet. 2002;111(4):366–75.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Members of PWS Cooperation Group of Rare Diseases Branch of Chinese Pediatric Society

Shan Xiao-Ou, The Second Affiliated Hospital of Wenzhou Medical University; Yang Yu, Jiangxi Provincial Children's Hospital; Zhang Hui-Feng, The Second Hospital of Hebei Medical University; Tian Zhi-Liang, Harbin Medical University the Second Affiliated Hospital; Sun Bo, Gansu Province Maternal and Child Health Hospital; Lu Mei, Women and Children's Hospital of Xiamen University School of Medicine; Cheng Ya-Ying, Hebei General Hospital; Yang Ying, Guiyang Province Maternal and Child Health Hospital; Yu Xiong-Ying, Jiangxi Province Children's Hospital; Zhang Jing, The First Affiliated Hospital of Zhengzhou University; Chen Xiao-Hong, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology; Yang Fan, West China 2nd University Hospital of Sichuan University; Ma Hong-Wei, Shengjing Hospital Affiliated to China Medical University; MireguIi. Maimaiti, First Affiliated Hospital of Xinjiang Medical University; Zhang Gai-Xiu, Shanxi Provincial Children's Hospital; Chen Xiao-Hong, Wuhan Children’s Hospital; Li Gui-Mie, Shandong Provincial Hospital; Tong Fan, Children's Hospital, Zhejiang University School of Medicine.

Zhejiang Expert Group for PWS

Zhi Ming-Qiang, Children's Hospital, Zhejiang University School of Medicine; Zhou Qiong, Hangzhou Children's Hospital; Gao Yuan and Wang Kan, Jinhua Municipal Central Hospital; Ying Xiao-Ming, Taizhou First People's Hospital; Zhang Jian-Ping, Ningbo Women and Children’s Hospital; Wang Chun-Lin, the 1st Hospital of Zhejiang University School of Medicine; Jiang Chun-Ming, Hangzhou 1st People’s Hospital of Zhejiang University School of Medicine; Xiao Rui, National Engineering Laboratory of Key Technologies for Birth Defect Prevention and Control were take part in discussing and revising this recommendation.

Funding

This work is supported by Zhejiang Provincial Program for the Cultivation of High-Level Innovative Health Talents (2014) and Zhejiang Provincial Department of Science and Technology (2021C03094).

Author information

Authors and Affiliations

Authors

Consortia

Contributions

ZC-C and JY-H were responded for this recommendation. DY-L, LF-H, ZH-W, MM-S, LX-Ping, LL, ZQ, WY, JY-H and ZC-C, were responded for data collection and manuscript writing. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Jiang Yong-Hui or Zou Chao-Chun.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang-Li, D., Fei-Hong, L., Hui-Wen, Z. et al. Recommendations for the diagnosis and management of childhood Prader-Willi syndrome in China. Orphanet J Rare Dis 17, 221 (2022). https://doi.org/10.1186/s13023-022-02302-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13023-022-02302-z

Keywords