Coverage Policy Manual
Policy #: 2013044
Category: Medicine
Initiated: November 2013
Last Review: March 2024
  Genetic Test: Epilepsy

Description:
Epilepsy is a disorder characterized by unprovoked seizures. It is a heterogenous condition that encompasses many different types of seizures and that varies in age of onset and severity. The common epilepsies, also called idiopathic epilepsy, are thought to have a complex, multifactorial genetic basis. There are also numerous rare epileptic syndromes that occur in infancy or early childhood and that may be caused by a single gene mutation. Genetic testing is commercially available for a large number of genetic mutations that may be related to epilepsy.
 
Epilepsy is defined as the occurrence of two or more unprovoked seizures. It is a common neurologic disorder, with approximate 3% of the population developing the disorder over their entire lifespan (Williams, 2013).
 
Epilepsy is heterogeneous in etiology and clinical expression and can be classified in a variety of ways. Most commonly, classification is done by the clinical phenotype, i.e., the type of seizures that occur. The International League Against Epilepsy (ILAE) developed the classification system that is widely used for clinical care and research purposes (Berg, 2010). Classification of seizures can also be done on the basis of age of onset: neonatal, infancy, childhood, and adolescent/adult.
 
Classification of Seizure Disorders by Type
 
    • Focal Onset (including aware and impaired awareness)
      • Motor onset
        • automatisms
        • atonica
        • clonic
        • epileptic spasmsa
        • hyperkinetic
        • myoclonic
        • tonic
      • Nonmotor Onset
        • autonomic
        • behavior arrest
        • cognitive
        • emotional
        • sensory
      • Focal to bilateral tonic-clonic
    • Generalized Onset
      • Motor
        • tonic-clonic
        • clonic
        • tonic
        • myoclonic
        • myoclonic-tonic-clonic
        • myoclonic-atonic
        • atonic
        • epileptic spams
      • Nonmotor (absense)
        • typical
        • atypical
        • myoclonic
        • eyelid myoclonia
    • Unknown Onset
      • Motor
        • tonic-clonic
        • epileptic spasms
      • Nonmotor
        • behavior arrest
 
Although genetic epilepsies are not discussed in the 2017 ILAE report, a 2010 ILAE report identified genetic epilepsies as conditions in which the seizures are a direct result of a known or presumed genetic defect(s) (Fisher, 2017; Berg, 2010). Genetic epilepsies are characterized by recurrent unprovoked seizures in patients who do not have demonstrable brain lesions or metabolic abnormalities. In addition, seizures are the core symptom of the disorder, and other symptomatology is not present, except as a direct result of seizures. This is differentiated from genetically determined conditions in which seizures are part of a larger syndrome, such as tuberous sclerosis, fragile X syndrome, or Rett syndrome.
 
The policy will focus on the category of genetic epilepsies in which seizures are the primary clinical manifestation. This category does not include syndromes that have multiple clinical manifestations, of which seizures may be one. Examples of syndromes that include seizures are Rett syndrome and tuberous sclerosis. Genetic testing for these syndromes will not be assessed in this policy but may be included in separate policies that specifically address genetic testing for that syndrome.
 
Genetic epilepsies can be further broken down by type of seizures. For example, genetic generalized epilepsy (GGE) refers to patients who have convulsive (grand mal) seizures, while genetic absence epilepsy (GAE) refers to patients with nonconvulsive (absence) seizures. The disorders are also sometimes classified by age of onset.
 
The category of genetic epilepsies includes a number of rare epilepsy syndromes that present in infancy or early childhood (Williams, 2013; Merwick, 2012). These syndromes are characterized by epilepsy as the primary manifestation, without associated metabolic or brain structural abnormalities. They are often severe and sometimes refractory to medication treatment. They may involve other clinical manifestations such as development delay and/or intellectual disability, which in many cases are thought to be caused by frequent uncontrolled seizures. In these cases, the epileptic syndrome may be classified as an epileptic encephalopathy, which is described by ILAE as disorders in which the epileptic activity itself may contribute to severe cognitive and behavioral impairments above and beyond what might be expected from the underlying pathology alone and that these can worsen over time. A partial list of severe early-onset syndromes is as follows:
   
    • Dravet syndrome
    • EFMR syndrome (epilepsy limited to females with mental retardation)
    • Nocturnal frontal lobe epilepsy
    • GEFS+ syndrome (genetic epilepsy with febrile seizures plus)
    • EIEE syndrome (early infantile epileptic encephalopathy with suppression burst)
    • West Syndrome
    • Ohtahara Syndrome
 
Dravet syndrome falls on a spectrum of SCN1A-related seizure disorders, which includes febrile seizures at the mild end to Dravet syndrome and intractable childhood epilepsy with generalized tonic-clonic seizures at the severe end. The spectrum may be associated with multiple seizure phenotypes, with a broad spectrum of severity; more severe seizure disorders may be associated with cognitive impairment, or deterioration (Miller, 2014). Ohtahara syndrome is a severe early-onset epilepsy syndrome characterized by intractable tonic spasms, other seizures, interictal electroencephalography abnormalities, and developmental delay. It may be secondary to structural abnormalities but has been associated with variants in the STXBP1 gene in rare cases. West syndrome is an early-onset seizure disorder associated with infantile spasms and the characteristic electroencephalography finding of hypsarrhythmia. Other seizure disorders presenting early in childhood may have a genetic component but are characterized by a more benign course, including benign familial neonatal seizures and benign familial infantile seizures.
 
Genetics of epilepsy
The common genetic epilepsies are primarily believed to involve multifactorial inheritance patterns. This follows the concept of a threshold effect, in which any particular genetic defect may increase the risk of epilepsy, but is not by itself causative (Petrovski, 2013). A combination of risk-associated genes, together with environmental factors, determines whether the clinical phenotype of epilepsy occurs. In this model, individual genes that increase the susceptibility to epilepsy have a relatively weak impact. Multiple genetic defects, and/or particular combination of genes, probably increase the risk by a greater amount. However, it is not well understood how many abnormal genes are required to exceed the threshold to cause clinical epilepsy, nor is it understood which combination of genes may increase the risk more than others.
 
Early-onset epilepsy syndromes may be single-gene disorders. Because of the small amount of research available, the evidence base for these rare syndromes is incomplete, and new mutations are currently being discovered frequently (Helbig, 2013).  
 
Some of the most common genes that have been associated with genetic epileptic syndromes are: KCNQ2 (Potassium channel), KCNQ3 (Potassium channel), SCN1A (Sodium channel α-subunit), SCN2A (Sodium channel α-subunit), SCN1B (Sodium channel α-subunit), GABRG2 (γ-aminobutyrate A-type subunit), GABRRA1 (γ-aminobutyrate A-type subunit), GABRD (γ-aminobutyrate subunit), CHRNA2 (Acetylcholine receptor α2 subunit), CHRNA4 (Acetylcholine receptor α4 subunit), CHRNB2 (Acetylcholine receptor β2 subunit), STXBP1 (Synaptic vesicle release), ARX (Homeobox gene), PCDH19 (Protocadherin cell-cell adhesion), EFHC1 (Calcium homeostasis), CACNB4 (Calcium channel subunit), CLCN2 (Chloride channel), and LGI1 (G-protein component) (Williams, 2013).
 
For the severe early epilepsy syndromes, the disorders most frequently reported to be associated with single-gene variants include generalized epilepsies with febrile seizures plus syndrome (associated with SCN1A, SCN1B, and GABRG2 variants), Dravet syndrome (associated with SCN1A variants, possibly modified by SCN9A variants), and epilepsy and intellectual disability limited to females (associated with PCDH19 variants). Ohtahara syndrome has been associated with variants in STXBP1 in cases where patients have no structural or metabolic abnormalities. West syndrome is often associated with chromosomal abnormalities or tuberous sclerosis or may be secondary to an identifiable infectious or metabolic cause, but when there is no underlying cause identified, it is thought to be due to a multifactorial genetic predisposition (Deprez, 2009).
 
Targeted testing for individual genes is available. Several commercial epilepsy genetic panels are also available. The number of genes included in the tests varies widely, from about 50 to over 450. The panels frequently include genes for other disorders such as neural tube defects, lysosomal storage disorders, cardiac channelopathies, congenital disorders of glycosylation, metabolic disorders, neurologic syndromes, and multisystemic genetic syndromes. Some panels are designed to be comprehensive while other panels target specific subtypes of epilepsy. Chambers et al reviewed comprehensive epilepsy panels from 7 U.S.-based clinical laboratories and found that between 1% and 4% of panel contents were genes not known to be associated with primary epilepsy (Chambers, 2016). Between 1% and 70% of the genes included on an individual panel were not on any other panel.
 
Treatment
The condition is generally chronic, requiring treatment with 1 or more medications to adequately control symptoms. Seizures can be controlled by antiepileptic medications in most cases, but some patients are resistant to medications, and further options such as surgery, vagus nerve stimulation, and/or the ketogenic diet can be used (Kwan, 2000).
 
Pharmacogenomics of epilepsy
Another area of interest for epilepsy is the pharmacogenomics of anti-epileptic medications. There are a wide variety of these medications, from numerous different classes.  The choice of medications, and the combinations of medications for patients who require treatment with more than one agent, is complex. Approximately one-third of patients are considered refractory to medications, defined as inadequate control of symptoms with a single medication (Cavalleri, 2011). These patients often require escalating doses and/or combinations of different medications. At present, selection of agents is driven by the clinical phenotype of seizures but has a large trial and error component in many refractory cases. The current focus of epilepsy pharmogenomics is in identifying genetic markers that identify patients who are likely to be refractory to the most common medications. This may lead to directed treatment that will result in a more efficient process for medication selection, and potentially more effective control of symptoms.
 
Of note, genotyping for the HLA-B*1502 allelic variant in patients of Asian ancestry, prior to considering drug treatment with carbamazepine due to risks of severe dermatologic reactions, is recommended by the U.S. Food and Drug Administration (FDA) labeling for carbamazepine (FDA, 2018).
 
Genetic testing for epilepsy
Commercial testing is available from numerous companies. Testing for individual genes is available for most, or all, or the genes listed above, as well as for a wider range of genes. Because of the large number of potential genes, panel testing is available from a number of genetic companies. These panels typically include large numbers of genes that have been implicated in diverse disorders.
 
GeneDx® offers a number of different epilepsy panels that have overlapping genes in varying combinations. The GeneDx® Comprehensive Epilepsy Panel lists 71 genes. They also offer a Childhood Onset epilepsy panel and an Infancy Panel. The GeneDx® infantile epilepsy panel includes the following 50 genes:
 
ADSL, ALDH7A1, ARX, ATP6AP2, CDKL5, CHRNA7, CLN3, CLN5, CLN6, CLN8, CNTNAP2, CTSD, FOLR1, FOXG1, GABRG2, GAMT, GRIN2A, KANSL1, KCNJ10, KCNQ2, KCNQ3, KCTD7, LIAS, MAGI2, MBD5, MECP2, MEF2C, MFSD8, NRXN1, PCDH19, PNKP, PNPO, POLG, PPT1, SCN1A, SCN1B, SCN2A, SCN8A, SLC25A22, SLC2A1, SLC9A6, SPTAN1, STXBP1, TBC1D24, TCF4, TPP1 (CLN2), TSC1, TSC2, UBE3A, ZEB2
 
The Courtagen epiSEEK® gene panel includes over 200 genes in its panel.
 
Emory Genetics® Epilepsy and Seizure Disorders panel offers testing of 123 different genetic mutations by next-generation sequencing.
 
Regulatory Status
Clinical laboratories may develop and validate tests in-house and market them as a laboratory service; laboratory-developed tests must meet the general regulatory standards of the Clinical Laboratory Improvement Amendments (CLIA). Commercially available genetic tests for epilepsy are available under the auspices of the CLIA. Laboratories that offer laboratory-developed tests must be licensed by the CLIA for high-complexity testing. To date, the FDA has chosen not to require any regulatory review of this test.
 
Coding
If the specific gene being tested has been codified in CPT, the appropriate CPT code would be reported. If the specific gene has not been codified in CPT, the unlisted molecular pathology code 81479 would be reported.
 
Category 1 CPT codes:
 
CPT code 81419
Epilepsy genomic sequence analysis panel, must include analyses for ALDH7A1, CACNA1A, CDKL5, CHD2, GABRG2, GRIN2A, KCNQ2, MECP2, PCDH19, POLG, PRRT2, SCN1A, SCN1B, SCN2A, SCN8A, SLC2A1, SLC9A6, STXBP1, SYNGAP1, TCF4, TPP1, TSC1, TSC2, and ZEB2) and 0232U (CSTB [cystatin B] [e.g., progressive myoclonic epilepsy type 1A, Unverricht Lundborg disease], full gene analysis, including small sequence changes in exonic and intronic regions, deletions, duplications, short tandem repeat [STR] expansions, mobile element insertions, and variants in non uniquely mappable regions) (Effective 1/1/2021)
 
The following is a list of some of the tests related to epilepsy that are listed under CPT Tier 2 codes:
 
Under CPT code 81401:
MT-TK (mitochondrially encoded tRNA lysine) (e.g., myoclonic epilepsy with ragged-red fibers [MERRF]), common variants (e.g., m.8344A>G, m.8356T>C)
 
Under CPT code 81403:
NHLRC1 (NHL repeat containing 1) (e.g., progressive myoclonus epilepsy), full gene sequence
 
Under CPT code 81404:
ARX (aristaless related homeobox) (e.g., X-linked lissencephaly with ambiguous genitalia, X-linked mental retardation), full gene sequence
 
EPM2A (epilepsy, progressive myoclonus type 2A, Lafora disease [laforin]) (e.g., progressive myoclonus epilepsy), full gene sequence
 
Under CPT code 81405:
CHRNA4 (cholinergic receptor, nicotinic, alpha 4) (e.g., nocturnal frontal lobe epilepsy), full gene sequence
 
CHRNB2 (cholinergic receptor, nicotinic, beta 2 [neuronal]) (e.g., nocturnal frontal lobe epilepsy), full gene sequence
 
GABRG2 (gamma-aminobutyric acid [GABA] A receptor, gamma 2) (e.g., generalized epilepsy with febrile seizures), full gene sequence
 
Under CPT code 81406:
ALDH7A1 (aldehyde dehydrogenase 7 family, member A1) (e.g., pyridoxine-dependent epilepsy), full gene sequence
 
CDKL5 (cyclin-dependent kinase-like 5) (e.g., early infantile epileptic encephalopathy), full gene sequence
 
EFHC1 (EF-hand domain [C-terminal] containing 1) (e.g., juvenile myoclonic epilepsy), full gene sequence
 
Under CPT code 81407:
SCN1A (sodium channel, voltage-gated, type 1, alpha subunit) (e.g., generalized epilepsy with epilepsy with febrile seizures), full gene sequence
 
Proprietary Laboratory Analysis Codes:
 
0232U CSTB (cystatin B) (e.g., progressive myoclonic epilepsy type 1A, Unverricht Lundborg disease), full gene analysis, including small sequence changes in exonic and intronic regions, deletions, duplications, short tandem repeat (STR) expansions, mobile element insertions, and variants in non uniquely mappable regions (Effective 1/1/2021)  
 
If a panel of tests that have not been codified in CPT is performed, code 81479 would be reported once.

Policy/
Coverage:
EFFECTIVE November 2015
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
Genetic testing of individuals with infantile- and early childhood-onset epilepsy syndromes in which epilepsy is the core clinical symptom meets member benefit certificate primary coverage criteria and is covered when ALL of the following criteria are met:
 
1. Epilepsy syndromes that present in infancy or early childhood, are severe, and are characterized by epilepsy as the primary manifestation, without associated metabolic or brain structural abnormalities.
2. There is a completed consult with a pediatric neurologist
3. There is a completed an EEG
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Genetic testing for epilepsy does not meet member benefit certificate primary coverage criteria for all other indications.
 
For members with contracts without primary coverage criteria genetic testing for epilepsy for all other indications is considered investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Note* This policy does not address the use of genotyping for the HLA-B*1502 allelic variant in patients of Asian ancestry prior to considering drug treatment with carbamazepine due to risks of severe dermatologic reactions (see policy 2007023). This testing is recommended by the Food and Drug Administration labeling for carbamazepine. (Administration FDA Label).
 
EFFECTIVE Prior to November 2015
 
Genetic testing for epilepsy does not meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes.
 
For members with contracts without primary coverage criteria, genetic testing for epilepsy is considered investigational.  Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Note: This policy statement covers testing for the common epilepsies where epilepsy is the only clinical manifestation and for which there is not a structural or metabolic defect predisposing to epilepsy as well as the rare epilepsy syndromes that present in infancy or early childhood, in which epilepsy is the core clinical symptom. This statement does not cover testing for syndromes that have a wider range of symptomatology, of which seizures may be one. Genetic testing for these syndromes may be specifically addressed in other  policies.

Rationale:
This policy was created in November 2013 with review of the literature through September 30, 2013.
 
The evaluation of a genetic test focuses on 3 main principles: 1) analytic validity (the technical accuracy of the test in detecting a mutation that is present or in excluding a mutation that is absent); 2) clinical validity (the diagnostic performance of the test [sensitivity, specificity, positive and negative predictive values] in detecting clinical disease); and 3) clinical utility (how the results of the diagnostic test will be used to change management of the patient and whether these changes in management lead to clinically important improvements in health outcomes)
 
The genetic epilepsies will be discussed in two categories: The rare epileptic syndromes that may be caused by a single-gene mutation and the common epilepsy syndromes that are thought to have a multifactorial genetic basis.
 
Rare Epilepsy Syndromes Associated with Single-Gene Mutations  
There are numerous rare syndromes that have seizures as their primary symptom.  These generally present in infancy or early childhood.  Many of them are thought to be caused by single-gene mutations.  The published literature on these syndromes generally consists of small cohorts of patients treated in tertiary care centers, with descriptions of genetic mutations that are detected in affected individuals.   
 
Some of these syndromes are listed below, with the putative causative genetic mutations:
 
Dravet syndrome (severe myoclonic epilepsy of infancy)
SCN1A
 
Early infantile epileptic encephalopathy
STXBP1
 
Generalized epilepsy with febrile seizures plus (GEFS+)
SCN1A, SCN2A, SCN1B, GABRG2
 
Epilepsy and mental retardation limited to females (EFMR)
PCDH19
 
Nocturnal frontal lobe epilepsy
CHRNA4, CHRNB2, CHRNA2
 
Analytic validity
These syndromes can be evaluated by single-gene analysis, which is generally performed by direct sequencing.  Direct sequencing is the gold standard for identifying specific mutations.  This testing method has an analytic validity of greater than 99%.  They can also be evaluated by genetic panel testing, which is generally done by next-generation sequencing.  This method has a lower analytic validity compared to direct sequencing, but is still considered to be very accurate, in the range of 95-99%.
 
Clinical validity
The literature on the clinical validity of these rare syndromes is limited, and for most syndromes, the clinical sensitivity and specificity is not defined.  Dravet syndrome is probably the most well-studied, and some evidence on the clinical validity of SCN1A mutations is available.  The clinical sensitivity has been reported to be in the 70-80% range. (8, 9) In one series of 64 patients, 51 (79%) were found to have SCN1A mutations (Mulley, 2006). The false-positive rate and the frequency of variants of uncertain significance, is not well characterized.   
 
For the other syndromes, the associations of the genetic mutations with the syndromes have been reported in case reports or very small numbers of patients.  Therefore, it is not possible to determine the clinical validity of the putative causative genetic mutations.
 
Clinical utility
One potential area of clinical utility for genetic testing may be in making a definitive diagnosis and avoiding further testing.  For most of these syndromes, the diagnosis is made by clinical criteria, and it is not known how often genetic testing leads to a definitive diagnosis when the diagnosis cannot be made by clinical criteria.
 
Another potential area of clinical utility may be in directing pharmacologic treatment.  For Dravet syndrome, the seizures are often refractory to common medications. Some experts have suggested that diagnosis of Dravet syndrome may therefore prompt more aggressive treatment, and/or avoidance of certain medications that are known to be less effective, such as carbamazepine (Mulley, 2006; Ottman, 2010). However, there are no studies that examine the frequency with which genetic testing leads to changes in medication management, and there are no studies that report on whether the efficacy of treatment directed by genetic testing is superior to efficacy of treatment without genetic testing.
 
Section Summary.  There are numerous rare epileptic syndromes which may be caused by single-gene mutations, but the evidence on genetic testing for these syndromes is insufficient to form conclusions on the clinical validity and clinical utility of genetic testing.  The syndrome with the greatest amount of evidence is Dravet syndrome.  The clinical sensitivity of testing patients with clinically defined Dravet syndrome is relatively high in small cohorts of patients.  There may be clinical utility in avoiding further testing and directing treatment, but there is only a small amount of evidence to suggest this and no evidence demonstrating that outcomes are improved.
 
Common Epilepsies
The common epilepsy syndromes, also known as idiopathic epilepsy, generally present in childhood, adolescence or early adulthood.  They include generalized or focal in nature, and may be convulsant (grand mal) or absence type.  They are generally thought to have a multifactorial genetic component.  
 
Analytic validity
The common epilepsies are generally evaluated by genetic panel testing. The larger, commercially available panels that include many mutations are generally performed by next-generation sequencing. This method has a lower analytic validity compared to direct sequencing, but is still considered to be very accurate, in the range of 95-99%.  Less commonly, deletion/duplication analysis may be performed; this method is also considered to have an analytic validity of greater than 95%.
 
Clinical validity
The literature on clinical validity includes many studies that report the association of various genetic variants with the common epilepsies.  There are a large number of case-control studies that compare the frequency of genetic variants in patients with epilepsy to the frequency in patients without epilepsy.  There are a smaller number of genome-wide association studies (GWAS) that evaluate the presence of single-nucleotide polymorphisms (SNPs) associated with epilepsy across the entire genome.  No studies were identified that reported the clinical sensitivity and specificity of genetic mutations in various clinically defined groups of patients with epilepsy.  In addition to these studies on the association of genetic variants with the diagnosis of epilepsy, there are numerous other studies that evaluate the association of genetic variants with pharmacogenomics of anti-epileptic medications.
 
Diagnosis of Epilepsy. The Epilepsy Genetic Association Database (epiGAD) published an overview of genetic association studies in 2010 (Tan, 2010). This review identified 165 case-control studies published between 1985 and 2008.  There were 133 studies that examined the association of 77 different genetic variants with the diagnosis of epilepsy. Approximately half of these studies (65/133) focused on patients with genetic generalized epilepsy.  Most of these studies had relatively small sample sizes, with a median of 104 cases (range 8-1,361) and 126 controls (range 22-1,390).  There were less than 200 case patients in 80% of the studies.  The majority of the studies did not show a statistically significant association. Using a cutoff of p<0.01 as the threshold for significance, there were 35 studies (21.2%) that reported a statistically significant association.  According to standard definitions for genetic association, all of the associations were in the weak to moderate range, with no associations reported that were considered strong.  
 
The EPICURE Consortium published one of the larger GWAS of genetic generalized epilepsy in 2012 (Epicure Consortium, 2012). This study included 3,020 patients with genetic generalized epilepsy (GGE) and 3,954 control patients, all of European ancestry.  A 2-stage approach was used, with a discovery phase and a replication phase, to evaluate a total of 4.56 million single-nucleotide polymorphisms (SNPs).  In the discovery phase, 40 candidate SNPs were identified that exceeded the significance for the screening threshold (1 x 10-5), although none of these reached the threshold defined as statistically significant for genome-wide association (1 x 10-8).  After stage 2 analysis, there were 4 SNPs identified that had suggestive associations with GGE on genes SCN1A, CHRM3, ZEB2, and NLE2F1.
 
A second GWAS with a relative large sample size of Chinese patients was also published in 2012 (Guo, 2012). Using a similar 2-stage methodology, this study evaluated 1,087 patients with epilepsy and 3,444 matched controls.  Two variants were determined to have the strongest association with epilepsy.  One of these was on the CAMSAP1L1 gene and the second was on the GRIK2 gene.  There were several other loci on genes that were suggestive of an association on genes that coded for neurotransmitters or other neuron function.
 
In contrast to the 2 studies, a GWAS published from the UK failed to show any robust associations of SNPs with partial epilepsy (Kasperaviciute, 2010). This study included 3,445 patients with partial epilepsies and 6,935 controls of European ancestry.  Using a threshold of an odds ratio greater than 1.3, the authors reported that no SNPs were identified that had a statistically significant association at that level.
 
In 2012, Heinzen et al. used whole exome sequencing to evaluate the association of genetic variants with genetic generalized epilepsy in 118 individuals with the disorder and 242 control patients of European origin (Heinzen, 2012). No variants were found that reached the statistical threshold for a statistical association.  From this initial data, the researchers selected 3,897 candidate genetic variants.  These variants were tested in a replication sample of 878 individuals with GGE and 1,830 controls. None of the tested variants showed a statistically significant association.
 
In addition to the individual studies, there are a number of meta-analyses that evaluate the association of particular genetic variants with different types of epilepsy.  Most of these have not shown a significant association.  For example, Cordoba et al. evaluated the association of SLC6A4 gene variants with temporal lobe epilepsy in a total of 991 case patients and 1,202 controls and failed to demonstrate a significant association on combined analysis (Cordoba, 2012).  Nurmohamed et al. performed a meta-analysis of 9 case-control studies that evaluated the association of the ABC1 gene polymorphisms with epilepsy (Nurmohamed, 2010). There were a total of 2,454 patients with epilepsy and 1,542 control patients.  No significant associations were found.  One meta-analysis that did report a significant association was published by Kauffman et al. In 2008 (Kauffman, 2008). This study evaluated the association of variants in the IL1B gene with temporal lobe epilepsy and febrile seizures, using data from 13 studies of 1,866 patients with epilepsy and 1,930 controls.  Combined analysis showed a significant relationship between one SNP (511T) and temporal lobe epilepsy, with a strength of association that was considered modest (odds ratio [OR]: 1.48, 95% confidence interval [CI]: 1.1-2.0, p=0.01).
 
Pharmacogenomics of anti-epileptic medications.  Numerous case-control studies report on the association of various genetic variants with response to medications in patients with epilepsy.  The epiGAD database identified 32 case-control studies of 20 different genes and their association with medication treatment (Tan, 2010). The most common comparison was between patients who were responders to medication and patients who were non-responders.  Some of the larger representative studies are discussed below.
 
Kwan et al. compared the frequency of SNPs on the SCN1A, SCN2A, and SCN3A genes in 272 drug responsive patients and 199 drug resistant patients (Kwan, 2008). A total of 27 candidate SNPs were evaluated, selected from a large database of previously identified SNPs.  There was one SNP identified on the SCN2A gene (rs2304016) that had a significant association with drug resistance (OR: 2.1, 95% CI: 1.2-3.7, p<0.007).
 
Jang et al. compared the frequency of variants on the SCN1A, SCN1B, and SCN2B genes in 200 patients with drug resistant epilepsy and 200 patients with drug responsive epilepsy (Jang, 2009). None of the individual variants tested showed a significant relationship with drug resistance. In further analysis of whether there were gene-gene interactions that were associated with drug resistance, the authors reported that there was a possible interaction of 2 variants, one on the SCN2A gene and the other on the SCN1B gene, that were of borderline statistical significance (p=0.055).
 
One meta-analysis evaluating pharmacogenomics was identified (Haerian, 2010). This study examined the association between SNPs on the ABCB1 gene and drug resistance in 3,231 drug resistant patients and 3,524 controls from 22 studies.  The authors reported no significant relationship between variants of this gene and drug resistance (combined OR: 1.06, 95% CI: 0.98-1.14, p=0.12).  There was also no significant association between on subgroup analysis by ethnicity.
 
Clinical utility
There is a lack of evidence on the clinical utility of genetic testing for the common genetic epilepsies.  Association studies are not sufficient evidence to determine whether genetic testing can improve the clinical diagnosis of GGE.  There are no studies that report the accuracy in terms of sensitivity, specificity, or predictive value; therefore it is not possible to determine the impact of genetic testing on diagnostic decision making.
 
The evidence on pharmacogenomics suggests that genetic factors may play a role in the pharmacokinetics of anti-epileptic medications.  However, this evidence does not provide guidance on how genetic information might be used to tailor medication management in ways that will improve efficacy, reduce adverse effects, or increase the efficiency of medication trials.
 
Section Summary.  The evidence on genetic testing for the common epilepsies is characterized by a large number of studies that evaluate associations of many different genetic variants with the various categories of epilepsy.  The evidence on clinical validity is not consistent in showing an association of any specific genetic mutation with any specific type of epilepsy.  Where associations have been reported, they are not of strong magnitude, and in most cases, have not been replicated independently or through the available meta-analyses. Because of the lack of established clinical validity, the clinical utility of genetic testing for the common epilepsies is also lacking.  
 
Summary    
Genetic testing for epilepsy covers a wide range of clinical syndromes and possible genetic defects.  For rare epilepsy syndromes, which may be caused by single-gene mutations, there is only a small body of research, which is insufficient to determine the clinical validity and clinical utility of genetic testing. There may be a potential role in differentiating these syndromes from the common epilepsies and from each other, and in improving the efficiency of the diagnostic work-up.  There also may be a potential role for genetic testing in identifying syndromes that are resistant to particular medications, and thereby directing treatment.  However, at the present time, the evidence is limited and the specific way in which genetic testing leads to improved outcomes is ill-defined.
 
For the common epilepsies, which are thought to have a complex, multifactorial basis, the role of specific genetic mutations remains uncertain. Despite a large body of literature of associations between genetic variants and common epilepsies, the clinical validity of genetic testing is poorly understood.  Published literature is characterized by weak and inconsistent associations, which have not been replicated independently or by meta-analyses. This literature does not permit conclusions on the clinical validity of genetic testing.  Because of the lack of conclusions on clinical validity, conclusions on clinical utility are also lacking.
 
For epilepsy pharmacogenomics, there are numerous studies that evaluate the associations of genetic variants with medication response.  This body of evidence also does not show consistent or strong relationships between genetic variants and response to medications. Therefore, the clinical utility of pharmacogenomics in epilepsy has not been demonstrated.
 
 
  
Regulatory Status
 
No U.S. Food and Drug Administration (FDA)-cleared genotyping tests were identified.  The available commercial genetic tests for epilepsy are offered as laboratory-developed tests.  Clinical laboratories may
 
2015 Update
A literature search conducted through October 2015 did not reveal any new information that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Clinical Input Received From Physician Specialty Societies and Academic Medical Centers
While the various physician specialty societies and academic medical centers may collaborate with and make recommendations during this process, through the provision of appropriate reviewers, input received does not represent an endorsement or position statement by the physician specialty societies or academic medical centers, unless otherwise noted.
 
In response to requests, input was received from 2 academic medical centers and 3 specialty societies, for a total of 6 reviewers while this policy was under review for 2015. The review was limited to input related to the use of genetic testing for infantile and early-childhood onset epileptic encephalopathies. There was consensus that genetic testing for early onset epileptic encephalopathies is medically necessary. Particular areas of clinical utility noted by reviewers included making specific treatment decisions in SCN1A-related epilepsies and avoiding other diagnostic tests and for reproductive planning for multiple types of early-onset epilepsies.
 
At the present time, evidence related to specific ways that genetic testing leads to improved outcomes is limited. Clinical input indicated strong support for the use of genetic testing in the evaluation of infantile- and early childhood- onset epilepsy syndromes associated with encephalopathy. The clinical utility of genetic testing occurs primarily when there is a positive test for a known pathogenic mutation. The presence of a pathogenic mutation may lead to targeted medication management, avoidance of other diagnostic tests, and/or informed reproductive planning.
 
Practice Guidelines and Position Statements
In 2010, the European Federation of Neurological Societies (EFNS) issued guidelines on the molecular diagnosis of channelopathies, epilepsies, migraine, stroke, and dementias.40 The guidelines made the following recommendations pertaining to epilepsy:
 
“There is good evidence to suggest that a thorough clinical and electrophysiological investigation may lead to the choice of gene to be tested in patients with periodic paralysis (level B). In myotonic disorders, it is recommended to first search for myotonic dystrophy and use clinical and electrophysiological phenotype characterization to guide for molecular genetic testing (level B). Molecular investigations are possible and may help in some cases to diagnose the condition but cannot be considered as a routine procedure with regard to the large number of different mutations in different genes. Furthermore, diagnosis can be made more easily by clinical and physiological investigation. One exception is the diagnosis of severe myoclonic epilepsy of infancy (SMEI), in which mutations are found in SCNIA in 80% of the patients (level B).”
 
2017 Update
 
A literature search conducted using the MEDLINE database through October 2017 did not reveal any new information that would prompt a change in the coverage statement.
 
Early-Onset Epilepsy And Epileptic Encephalopathies
 
Moller et al (2016) reported the testing yield with an epilepsy gene panel including 46 genes in a cohort of 216 consecutive patients referred for genetic testing with epileptic encephalopathies phenotypes or familial epilepsy (Moller, 2016). The patients ranged in age from 2 weeks to 74 years; the majority (52%) had epileptic encephalopathies. The criterion for including a gene in the panel was that it had been reported more than once in patients with monogenic epilepsies as of January 2014. Overall, a presumed disease-causing variant was found in 49 (23%) patients and a variant of uncertain significance (VUS) was found in 3%. The yield was highest in patients with epileptic encephalopathies (32%) and neonatal-onset epilepsies (57%). Variants were found in 19 genes, including SCN1A, STXBP1, CDKL5, SCN2A, SCN8A, GABRA1, KCNA2, and STX1B.
 
Trump et al (2016) also reported the yield of a gene panel including 46 genes in 400 patients with early-onset seizure disorders and/or severe developmental delay who were referred for gene panel testing in the United Kingdom (Trump, 2016)  Patients with major structural brain malformations or clinically significant copy number defects on microarray were not included. Authors reported that genes were included in the panel if they had been “established” as causes of early-onset seizures and/or severe developmental delay in patients without frequent major structural brain anomalies. Approximately half of the included genes overlapped with genes on the panel from Moller et al. Genes were added to the panel over time so that the original panel used in the first 48 patients included 29 genes, a second panel used in 94 patients included 39 genes, and the final panel used in the remaining 258 patients included 46 genes. Variants were found in 21 genes, most commonly SCN2A, CDKL5, KCNQ2, SCN8A, FOXG1, MECP2, SCNA1, STXBP1, KCNT1, PCDH19, and TCF4.
 
Prediction of Sudden Unexplained Death in Epilepsy
Coll et al (2016) evaluated the use of a custom resequencing panel including genes related to sudden death, epilepsy, and SUDEP in a cohort of 14 patients with focal or generalized epilepsy and a personal or family history of SUDEP, including 2 postmortem cases (Coll, 2016). In 4 cases, rare variants were detected with complete segregation in the SCN1A, FBN1, HCN1, SCN4A, and EFHC1 genes, and in 1 case a rare variant in KCNQ1 with an incomplete pattern of inheritance was detected. New potential candidate genes for SUDEP were detected: FBN1, HCN1, SCN4A, EFHC1, CACNA1A, SCN11A, and SCN10A. Bagnall et al (2016) performed an exome-based analysis of rare variants related to cardiac arrhythmia, respiratory control, and epilepsy to search for genetic risk factors in 61 SUDEP cases compared to 2936 controls (Bagnall, 2016). Mean epilepsy onset of the SUDEP cases was 10 years and mean age at death was 28 years. De novo variants, previously reported pathogenic variants, or candidate pathogenic variants were identified in 28 (46%) of 61 SUDEP cases. Four (7%) SUDEP cases had variants in common genes responsible for long QT syndrome and a further 9 (15%) cases had candidate pathogenic variants in dominant cardiac arrhythmia genes. Fifteen (25%) cases had variants or candidate pathogenic variants in epilepsy genes; 6 cases had a variant in DEPDC5. DEPDC5 (p=0.00015) and KCNH2 (p=0.0037) were highly associated with SUDEP. However, using a rare variant collapsing analysis, no gene reached criteria for genome-wide significance.
 
2018 Update
A literature search was conducted through October 2018.  There was no new information identified that would prompt a change in the coverage statement.  
 
2019 Update
Annual policy review completed with a literature search using the MEDLINE database through October 2019. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Esterhuizen et al analyzed data from 22 South African infants with provisional diagnoses of Dravet syndrome (DS) who underwent targeted resequencing of DS-associated genes (Esterhuizen, 2018). Disease-causing variants (SCN1A = 9, PCDH19 = 1) were identified in 10 children (45.5%), and results suggested that a clinical DS risk score of >6 and seizure onset before age 6 months were highly predictive of SCN1Aassociated DS. For 10 of the 12 variant-negative children, clinical reassessment resulted in a revised diagnosis. No limitations to the analysis were reported.
 
 Peng et al published an analysis of 273 pediatric patients with drug-resistant epilepsy who underwent genetic testing using whole exome sequencing (WES; n=74), epilepsy-related gene panel testing (n=141), or clinical WES gene panel testing (n=58) (Peng, 2018). Ninety-three likely disease-causing mutations in 33 genes were identified in 86 individuals (31.5%). The most frequently mutated genes were SCN1A (24.4%), TSC2 (8.1%), SCN8A (5.8%), CDKL5 (5.8%), KCNMA1 (4.6%), TSC1 (4.6%), KCNQ2 (3.5%), MECP2 (3.5%), PCDH19 (3.5%), and STXBP1(3.5%). Of the 34 individuals who accepted corrective therapy according to their mutant genes, 52.9% became seizure-free and 38.2% achieved seizure reduction. No limitations to the analysis were reported.
 
Hesse et al published a retrospective analysis of 305 patients (age range <1–69 years old with 88% <18 years old) referred for genetic testing with a targeted epilepsy panel between 2014 and 2016 (Hesse, 2018).34 Positive yield was 15.1%, with pathogenic, likely pathogenic, predicted deleterious mutations identified in 46 individuals. Twenty-nine distinct genes were present, and known pathogenic variants were identified in 7 genes (BRAF, DPYD, GABRG2, PAX6, SCN1A, SLC2A1, and SLC46A1). No limitations to the analysis were reported.  
 
Lindy et al published an industry sponsored analysis of 8565 consecutive individuals with epilepsy and/or neurodevelopmental disorders who underwent genetic testing with multigene panels (Lindy, 2018). Positive results were reported in 1315 patients (15.4%), and, of 22 genes with high positive yield, SCN1A (24.8%) and KCNQ2 (13.2%) accounted for the greatest number of positive findings. Results found 14 distinct genes with recurrent pathogenic or likely pathogenic (P/LP) variants (most commonly in MECP2, KCNQ2, SCN1A, SCN2A, STXBP1, and PRRT2). Greater than 30% of positive cases had parental testing performed; all variants found in CDKL5, STXBP1, SCN8A, GABRA1, and FOXG1 were de novo, however, 85.7% of variants in PRRT2 were inherited. No P/LP variants were found in ATP6AP2, CACNB4, CHRNA2, DNAJC5, EFHC1, MAGI2, and SRPX2. No limitations to the analysis were reported.
 
Miao et al published an analysis of 141 Chinese patients under 14 years of age with epilepsy who underwent genotype and phenotype analysis using an epilepsy-associated gene panel between 2015 and 2017 (Miao, 2018). Certain diagnoses were obtained in 39 probands (27.7%); these causative variants were related to 21 genes. The most frequently mutated gene was SCN1A (5.6%), but others included KCNQ2, KCNT1, PCDH19, STXBP1, SCN2A, TSC2, and PRRT2. The treatments for 18 patients (12.8%) were altered based on their genetic diagnosis and on genotype-phenotype analysis. No limitations to the analysis were reported.
 
Butler et al published a retrospective analysis of epilepsy patients screened using a 110-gene panel between 2013 and 2016; 339 unselected individuals (age range 2.5 months to 74 years, with more than 50% <5 years old) were included (Butler, 2017). Pathogenic and likely pathogenic variants were identified in 62 patients (18%), and another 21 individuals (6%) had potentially causative variants. SCN1A (n=15) and KCNQ2 (n=10) were the frequently identified potentially causative variants. However, other genes in which variants were identified in multiple individuals included CDKL5, SCN2A, SCN8A, SCN1B, STXBP1, TPP1, PCDH19, CACNA1A, GABRA1, GRIN2A, SLC2A1, and TSC2. The study was limited by the lack of clinical information available for approximately 20% of participants.
 
2020 Update
Annual policy review completed with a literature search using the MEDLINE database through October 2020. No new literature was identified that would prompt a change in the coverage statement.
 
2021 Update
Annual policy review completed with a literature search using the MEDLINE database through October 2021. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
Another single-center retrospective study by Hoelz et al. described the effect of next-generation sequencing on clinical decision-making among children with epilepsy (Hoelz, 2020). Testing was performed a mean of 3.6 years after symptom onset. Most of the patients had epileptic encephalopathy (40%) followed by focal epilepsy (33%) and generalized seizures (18%). Sixteen patients (18%) who underwent testing had a pathogenic or likely pathogenic gene identified. Subsequently, 10 of these 16 patients (63%) had changes in their clinical management, including medications (n=7), diagnostic testing (n=8), or avoiding future surgical procedures (n=2).
 
Alsubaie et al. evaluated the diagnostic yield of whole exome sequencing among 420 patients at a single center in Saudi Arabia (Alsubaie, 2020). Epilepsy was the reason for testing in 15.4% (n=65) patients. Whole exome sequencing confirmed the diagnosis of epilepsy in 14 patients (positive yield of 21.5%) with variants in the following genes: ARID1B, UGDH, KCNQ2, PAH, PARS2, ARHGEF9, CNA2, CASK, SLC23A3, TBCD, QARS, CBL, GABRB2, and SUOX. Genetic test results were inconclusive in 15 of the 65 patients with epilepsy (23%). Thirty patients with negative whole exome sequencing results underwent comparative genomic hybridization, which identified 4 additional variants (positive yield of 13.3%).
 
Johannsen et al. published a cohort study of 200 adult patients (range 18 to 80 years) with epilepsy who were referred for genetic testing between 2013 and 2018 in Denmark (Johannsen, 2020). Most of the patients (91%) also had intellectual disability. Various gene panels (range 45 to 580 genes) were used. A genetic cause of epilepsy was identified in 23% of patients (n=46). Pathogenic variants were found in 22 genes (SCN1A, KCNT1, STXBP1, CDKL5, CHD2, PURA, ATP6V1A, DCX, GABRB3, GABRG2, GRIN2A, HNRNPU, IQSEC2, KCNA2, KIAA2022, MECP2, MEF2C, MTOR, IPF2PBL, PCDH19, SCN8A, SLC2A1, SYNGAP1, and IRF2BPL). Among the 46 patients who received a diagnosis, variants in the SCN1A gene were most prevalent (36%). A change in management occurred in 11 patients after diagnosis, which led to improved seizure control and/or cognitive function.
 
Minardi et al. published a single-center analysis of 71 adult patients (age range: 21 to 65 years) with developmental and epileptic encephalopathies of unknown etiology who underwent whole exome sequencing (Minardi, 2020). Almost all patients (90.1%) had prior negative genetic tests. The analysis identified 24 variants that were considered pathogenic or likely pathogenic. The variants were: DYNC1, ZBTB20, CACNA1, DYRK1A, ANKRD11, GABRG2, KCNB1, KCNH5, SCN1A, GABRB2, YWHAG, STXBP1, PRODH, LAMB1, PNKP, APC2, RARS2, KIAA2022, and SMC1A. No clinical characteristics were significantly different between patients with pathogenic variants and patients with variants of unknown clinical significance; however, sample sizes were small. In half of the diagnosed cases (n=9), clinical management changed after diagnosis, including medication selection, additional testing, and reproduction-related decisions.
 
In 2017, recommendations were published from a consensus panel of 14 physicians and 5 family members/caregivers of patients with Dravet syndrome (Wirrell, 2017). There was strong consensus among panel members that genetic testing should be completed in all patients with clinical suspicion for Dravet syndrome since this can lead to earlier diagnosis. Options for testing include SCN1A sequencing followed by testing for deletions and duplications if sequencing is negative, or epilepsy gene panel testing, with no consensus among panel members about which option is superior. There was strong consensus that epilepsy gene panel testing is preferred to SCN1A testing if the clinical presentation is less clear or if the patient has atypical features, and that karyotyping is not needed. The panel did not reach consensus about the utility of chromosomal microarray in patients with suspected Dravet syndrome (72.2% agreed, 22.2% disagreed, 5.6% did not know) and concluded that this test can be considered. Based on strong consensus, the panel recommended genetic testing in the following circumstances among children with normal development, seizures of unknown etiology, and no evidence of causal lesion in the brain: infants with at least 2 prolonged focal febrile seizures, or children aged 1 to 3 years with at least one prolonged febrile seizure before 18 months of age or myoclonic or atypical absence seizures that are refractory to at least one antiepileptic medication. Infants who experience a single prolonged focal or generalized convulsion do not require genetic testing (strong consensus), but this can be considered in children aged 1 to 3 years who experience multiple brief episodes of febrile seizure activity before 18 months of age or myoclonic or atypical absence seizures that do not respond to antiepileptic medication (moderate consensus). The panel had moderate consensus about the role of genetic testing (epilepsy gene panel) in teens and adults without congenital dysmorphism who have seizure activity resistant to antiepileptic medication and lack an early life history.
 
2022 Update
Annual policy review completed with a literature search using the MEDLINE database through October 2022. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
A number of studies have reported on the genetic testing yield in cohorts of pediatric patients with epilepsy, typically in association with other related symptoms. Below are some examples of genetic testing diagnostic yield in children with epileptic encephalopathy:
    • A study by Jiang et al consisted of 221 children with epilepsy (Jiang, 2021). Whole exome sequencing was done. Diagnostic yield:
      • 64.5% of patients with epilepsy and developmental delay/intellectual disability; 18.9% of patients with only epilepsy (p<.001)
      • 48 of 87 variants detected were novel (55.2%)
      • Genes with novel variants: NCL, SEPHS2, PA2G4, SLC35G2, MYO1C, GPR158, and POU3F1
 
    • A study by Kim et al consisted of 59 patients with infantile-onset epilepsy and prior negative targeted gene panel testing (Kim, 2021). Whole exome sequencing was done. Diagnostic yield:
      • 8% more patients than with targeted gene panel testing
      • Genes with pathogenic or likely pathogenic variants: FARS2, YWHAG, KCNC1, DYRK1A, SMC1A, PIGA, OGT, and FGF12
      • Genes newly associated with epilepsy: YWHAG, KCNC1, and FGF12
 
    • A study by Palmer et al consisted of 30 patients with developmental and epileptic encephalopathies with prior negative genetic testing (Palmer, 2021). Whole exome sequencing was done. Diagnostic yield:
      • 53% in 15 patients with prior exome sequencing; 20% (3 of 15) had complex structural variants
      • 68% in 15 patients with prior multigene panel testing
 
    • A study by Salinas et al consisted of 55 patients with developmental and epileptic encephalopathies with prior negative genetic testing (Salinas, 2021). Targeted multigene panel testing and whole exome sequencing was done. Diagnostic yield:
      • 38% at baseline
      • 53% after an average of 29 months using new literature
      • Genes with novel variants: CHD2, COL4A1, FOXG1, GABRA1, GRIN2B, HNRNPU, KCNQ2, MECP2, PCDH19, SCN1A, SCN2A, SCN8A, SLC6A1, STXBP1, and WWOX
 
    • A study by Sun et al consisted of 73 infants with epileptic encephalopathies including West syndrome and Dravet syndrome (Sun, 2021). Whole exome sequencing was done. Diagnostic yield:
      • 46.6% (most commonly SCN1A variants)
      • Genes with novel variants: CACNA1E and WDR26
 
McKnight et al. conducted targeted gene panel testing (range, 89 to 189 genes) using next-generation sequencing in a cohort of 2008 adults with epilepsy (McKnight, 2021). Diagnosis occurred in 10.9% of patients, and 55.5% of these diagnoses led to changes in clinical management. Diagnostic yield was highest among individuals who first experienced seizure activity during infancy (29.6%) and among females with developmental delay or intellectual disability (19.6%). Patients with treatment-resistant epilepsy had a diagnostic yield of 13.5% and 57.4% of diagnoses led to changes in clinical management. The most common genes associated with a diagnosis were SCN1A and MECP2. The most common genes associated with changes in clinical management were SCN1A, DEPDC5, PRRT2, PCDH19, and TSC1. Nondiagnostic and negative genetic findings were common (70.1% and 19.0%, respectively).
 
Lin et al. conducted a prospective study of 96 children (age <2 years) with epilepsy and neurodevelopmental disability (Lin, 2021). A genetic cause of epilepsy was present in 28 children, while the remaining 68 children did not have an identified genetic cause. The incidence of drug-resistant epilepsy was 42.8% in patients with a genetic cause and 13.2% in patients without a genetic cause. Risk of drug-resistant epilepsy was significantly higher in the genetic group compared to the non-genetic group (adjusted OR, 6.50; 95% CI, 2.15 to 19.6; p=.03). Specific genes associated with drug-resistant epilepsy included TBC1D24, SCN1A, PIGA, PPP1CB, and SZT2.
 
2023 Update
Annual policy review completed with a literature search using the MEDLINE database through October 2023. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
The study by Scheffer et al included 103 children and infants with developmental and epileptic encephalopathies. Singleton exome sequencing was done with the following results: 35% of patients with genetic etiology, 29% had pathogenic or likely pathogenic variants, 38% had variants of unknown significance, and 33% were negative on exome analysis. KCNQ2, CDKL5, SCN1A, and STXBP1 were the most frequently identified genes (Scheffer, 2023). Thirteen of 36 patients with a known genetic cause for their condition had management implications. These included treatment for the underlying biochemical abnormality (1 patient with SLC2A1), choice of antiseizure medication (4 patients with KCNQ2, 3 with SCN1A, 2 with SCN8A, and 1 with SCN2A), choice of other medication (1 patient with ATP1A3), and screening for disease-related complications (1 patient with COL4A1).
 
2024 Update
Annual policy review completed with a literature search using the MEDLINE database through February  2024. No new literature was identified that would prompt a change in the coverage statement. The key identified literature is summarized below.
 
In 2022, the National Society of Genetic Counselors published a practice guideline on genetic testing and counseling for unexplained epilepsies (Smith, 2023).The Society made the following relevant recommendations:
 
  • "We strongly recommend that individuals with unexplained epilepsy be offered genetic testing, without limitation of age.
 
  • We strongly recommend comprehensive, multi-gene testing, such as exome/genome sequencing or multi-gene panel as a first-tier test.
 
  • We conditionally recommend exome/genome sequencing over multi-gene panel as the first-tier test.
 
  • The multi-gene panel should have a minimum of 25 genes and include copy number analysis."

CPT/HCPCS:
0232UCSTB (cystatin B) (eg, progressive myoclonic epilepsy type 1A, Unverricht Lundborg disease), full gene analysis, including small sequence changes in exonic and intronic regions, deletions, duplications, short tandem repeat (STR) expansions, mobile element insertions, and variants in non uniquely mappable regions
81401Molecular pathology procedure, Level 2 (eg, 2-10 SNPs, 1 methylated variant, or 1 somatic variant [typically using nonsequencing target variant analysis], or detection of a dynamic mutation disorder/triplet repeat) ABCC8 (ATP-binding cassette, sub-family C [CFTR/MRP], member 8) (eg, familial hyperinsulinism), common variants (eg, c.3898-9G&gt;A [c.3992-9G&gt;A], F1388del) ABL1 (ABL proto-oncogene 1, non-receptor tyrosine kinase) (eg, acquired imatinib resistance), T315I variant ACADM (acyl-CoA dehydrogenase, C-4 to C-12 straight chain, MCAD) (eg, medium chain acyl dehydrogenase deficiency), commons variants (eg, K304E, Y42H) ADRB2 (adrenergic beta-2 receptor surface) (eg, drug metabolism), common variants (eg, G16R, Q27E) APOB (apolipoprotein B) (eg, familial hypercholesterolemia type B), common variants (eg, R3500Q, R3500W) APOE (apolipoprotein E) (eg, hyperlipoproteinemia type III, cardiovascular disease, Alzheimer disease), common variants (eg, *2, *3, *4) CBFB/MYH11 (inv(16)) (eg, acute myeloid leukemia), qualitative, and quantitative, if performed CBS (cystathionine-beta-synthase) (eg, homocystinuria, cystathionine beta-synthase deficiency), common variants (eg, I278T, G307S) CFH/ARMS2 (complement factor H/age-related maculopathy susceptibility 2) (eg, macular degeneration), common variants (eg, Y402H [CFH], A69S [ARMS2]) DEK/NUP214 (t(6;9)) (eg, acute myeloid leukemia), translocation analysis, qualitative, and quantitative, if performed E2A/PBX1 (t(1;19)) (eg, acute lymphocytic leukemia), translocation analysis, qualitative, and quantitative, if performed EML4/ALK (inv(2)) (eg, non-small cell lung cancer), translocation or inversion analysis ETV6/RUNX1 (t(12;21)) (eg, acute lymphocytic leukemia), translocation analysis, qualitative, and quantitative, if performed EWSR1/ATF1 (t(12;22)) (eg, clear cell sarcoma), translocation analysis, qualitative, and quantitative, if performed EWSR1/ERG (t(21;22)) (eg, Ewing sarcoma/peripheral neuroectodermal tumor), translocation analysis, qualitative, and quantitative, if performed EWSR1/FLI1 (t(11;22)) (eg, Ewing sarcoma/peripheral neuroectodermal tumor), translocation analysis, qualitative, and quantitative, if performed EWSR1/WT1 (t(11;22)) (eg, desmoplastic small round cell tumor), translocation analysis, qualitative, and quantitative, if performed F11 (coagulation factor XI) (eg, coagulation disorder), common variants (eg, E117X [Type II], F283L [Type III], IVS14del14, and IVS14+1G&gt;A [Type I]) FGFR3 (fibroblast growth factor receptor 3) (eg, achondroplasia, hypochondroplasia), common variants (eg, 1138G&gt;A, 1138G&gt;C, 1620C&gt;A, 1620C&gt;G) FIP1L1/PDGFRA (del[4q12]) (eg, imatinib-sensitive chronic eosinophilic leukemia), qualitative, and quantitative, if performed FLG (filaggrin) (eg, ichthyosis vulgaris), common variants (eg, R501X, 2282del4, R2447X, S3247X, 3702delG) FOXO1/PAX3 (t(2;13)) (eg, alveolar rhabdomyosarcoma), translocation analysis, qualitative, and quantitative, if performed FOXO1/PAX7 (t(1;13)) (eg, alveolar rhabdomyosarcoma), translocation analysis, qualitative, and quantitative, if performed FUS/DDIT3 (t(12;16)) (eg, myxoid liposarcoma), translocation analysis, qualitative, and quantitative, if performed GALC (galactosylceramidase) (eg, Krabbe disease), common variants (eg, c.857G&gt;A, 30-kb deletion) GALT (galactose-1-phosphate uridylyltransferase) (eg, galactosemia), common variants (eg, Q188R, S135L, K285N, T138M, L195P, Y209C, IVS2-2A&gt;G, P171S, del5kb, N314D, L218L/N314D) H19 (imprinted maternally expressed transcript [non-protein coding]) (eg, Beckwith-Wiedemann syndrome), methylation analysis IGH@/BCL2 (t(14;18)) (eg, follicular lymphoma), translocation analysis; single breakpoint (eg, major breakpoint region [MBR] or minor cluster region [mcr]), qualitative or quantitative (When both MBR and mcr breakpoints are performed, use 81278) KCNQ1OT1 (KCNQ1 overlapping transcript 1 [non-protein coding]) (eg, Beckwith-Wiedemann syndrome), methylation analysis LINC00518 (long intergenic non-protein coding RNA 518) (eg, melanoma), expression analysis LRRK2 (leucine-rich repeat kinase 2) (eg, Parkinson disease), common variants (eg, R1441G, G2019S, I2020T) MED12 (mediator complex subunit 12) (eg, FG syndrome type 1, Lujan syndrome), common variants (eg, R961W, N1007S) MEG3/DLK1 (maternally expressed 3 [non-protein coding]/delta-like 1 homolog [Drosophila]) (eg, intrauterine growth retardation), methylation analysis MLL/AFF1 (t(4;11)) (eg, acute lymphoblastic leukemia), translocation analysis, qualitative, and quantitative, if performed MLL/MLLT3 (t(9;11)) (eg, acute myeloid leukemia), translocation analysis, qualitative, and quantitative, if performed MT-ATP6 (mitochondrially encoded ATP synthase 6) (eg, neuropathy with ataxia and retinitis pigmentosa [NARP], Leigh syndrome), common variants (eg, m.8993T&gt;G, m.8993T&gt;C) MT-ND4, MT-ND6 (mitochondrially encoded NADH dehydrogenase 4, mitochondrially encoded NADH dehydrogenase 6) (eg, Leber hereditary optic neuropathy [LHON]), common variants (eg, m.11778G&gt;A, m.3460G&gt;A, m.14484T&gt;C) MT-ND5 (mitochondrially encoded tRNA leucine 1 [UUA/G], mitochondrially encoded NADH dehydrogenase 5) (eg, mitochondrial encephalopathy with lactic acidosis and stroke-like episodes [MELAS]), common variants (eg, m.3243A&gt;G, m.3271T&gt;C, m.3252A&gt;G, m.13513G&gt;A) MT-RNR1 (mitochondrially encoded 12S RNA) (eg, nonsyndromic hearing loss), common variants (eg, m.1555A&gt;G, m.1494C&gt;T) MT-TK (mitochondrially encoded tRNA lysine) (eg, myoclonic epilepsy with ragged-red fibers [MERRF]), common variants (eg, m.8344A&gt;G, m.8356T&gt;C) MT-TL1 (mitochondrially encoded tRNA leucine 1 [UUA/G]) (eg, diabetes and hearing loss), common variants (eg, m.3243A&gt;G, m.14709 T&gt;C) MT-TL1 MT-TS1, MT-RNR1 (mitochondrially encoded tRNA serine 1 [UCN], mitochondrially encoded 12S RNA) (eg, nonsyndromic sensorineural deafness [including aminoglycoside-induced nonsyndromic deafness]), common variants (eg, m.7445A&gt;G, m.1555A&gt;G) MUTYH (mutY homolog [E. coli]) (eg, MYH-associated polyposis), common variants (eg, Y165C, G382D) NOD2 (nucleotide-binding oligomerization domain containing 2) (eg, Crohn's disease, Blau syndrome), common variants (eg, SNP 8, SNP 12, SNP 13) NPM1/ALK (t(2;5)) (eg, anaplastic large cell lymphoma), translocation analysis PAX8/PPARG (t(2;3) (q13;p25)) (eg, follicular thyroid carcinoma), translocation analysis PRAME (preferentially expressed antigen in melanoma) (eg, melanoma), expression analysis PRSS1 (protease, serine, 1 [trypsin 1]) (eg, hereditary pancreatitis), common variants (eg, N29I, A16V, R122H) PYGM (phosphorylase, glycogen, muscle) (eg, glycogen storage disease type V, McArdle disease), common variants (eg, R50X, G205S) RUNX1/RUNX1T1 (t(8;21)) (eg, acute myeloid leukemia) translocation analysis, qualitative, and quantitative, if performed SS18/SSX1 (t(X;18)) (eg, synovial sarcoma), translocation analysis, qualitative, and quantitative, if performed SS18/SSX2 (t(X;18)) (eg, synovial sarcoma), translocation analysis, qualitative, and quantitative, if performed VWF (von Willebrand factor) (eg, von Willebrand disease type 2N), common variants (eg, T791M, R816W, R854Q)
81403Molecular pathology procedure, Level 4 (eg, analysis of single exon by DNA sequence analysis, analysis of &gt;10 amplicons using multiplex PCR in 2 or more independent reactions, mutation scanning or duplication/deletion variants of 2-5 exons) ANG (angiogenin, ribonuclease, RNase A family, 5) (eg, amyotrophic lateral sclerosis), full gene sequence ARX (aristaless-related homeobox) (eg, X-linked lissencephaly with ambiguous genitalia, X-linked mental retardation), duplication/deletion analysis CEL (carboxyl ester lipase [bile salt-stimulated lipase]) (eg, maturity-onset diabetes of the young [MODY]), targeted sequence analysis of exon 11 (eg, c.1785delC, c.1686delT) CTNNB1 (catenin [cadherin-associated protein], beta 1, 88kDa) (eg, desmoid tumors), targeted sequence analysis (eg, exon 3) DAZ/SRY (deleted in azoospermia and sex determining region Y) (eg, male infertility), common deletions (eg, AZFa, AZFb, AZFc, AZFd) DNMT3A (DNA [cytosine-5-]-methyltransferase 3 alpha) (eg, acute myeloid leukemia), targeted sequence analysis (eg, exon 23) EPCAM (epithelial cell adhesion molecule) (eg, Lynch syndrome), duplication/deletion analysis F8 (coagulation factor VIII) (eg, hemophilia A), inversion analysis, intron 1 and intron 22A F12 (coagulation factor XII [Hageman factor]) (eg, angioedema, hereditary, type III; factor XII deficiency), targeted sequence analysis of exon 9 FGFR3 (fibroblast growth factor receptor 3) (eg, isolated craniosynostosis), targeted sequence analysis (eg, exon 7) (For targeted sequence analysis of multiple FGFR3 exons, use 81404) GJB1 (gap junction protein, beta 1) (eg, Charcot-Marie-Tooth X-linked), full gene sequence GNAQ (guanine nucleotide-binding protein G[q] subunit alpha) (eg, uveal melanoma), common variants (eg, R183, Q209) Human erythrocyte antigen gene analyses (eg, SLC14A1 [Kidd blood group], BCAM [Lutheran blood group], ICAM4 [Landsteiner-Wiener blood group], SLC4A1 [Diego blood group], AQP1 [Colton blood group], ERMAP [Scianna blood group], RHCE [Rh blood group, CcEe antigens], KEL [Kell blood group], DARC [Duffy blood group], GYPA, GYPB, GYPE [MNS blood group], ART4 [Dombrock blood group]) (eg, sickle-cell disease, thalassemia, hemolytic transfusion reactions, hemolytic disease of the fetus or newborn), common variants HRAS (v-Ha-ras Harvey rat sarcoma viral oncogene homolog) (eg, Costello syndrome), exon 2 sequence KCNC3 (potassium voltage-gated channel, Shaw-related subfamily, member 3) (eg, spinocerebellar ataxia), targeted sequence analysis (eg, exon 2) KCNJ2 (potassium inwardly-rectifying channel, subfamily J, member 2) (eg, Andersen-Tawil syndrome), full gene sequence KCNJ11 (potassium inwardly-rectifying channel, subfamily J, member 11) (eg, familial hyperinsulinism), full gene sequence Killer cell immunoglobulin-like receptor (KIR) gene family (eg, hematopoietic stem cell transplantation), genotyping of KIR family genes Known familial variant not otherwise specified, for gene listed in Tier 1 or Tier 2, or identified during a genomic sequencing procedure, DNA sequence analysis, each variant exon (For a known familial variant that is considered a common variant, use specific common variant Tier 1 or Tier 2 code) MC4R (melanocortin 4 receptor) (eg, obesity), full gene sequence MICA (MHC class I polypeptide-related sequence A) (eg, solid organ transplantation), common variants (eg, *001, *002) MT-RNR1 (mitochondrially encoded 12S RNA) (eg, nonsyndromic hearing loss), full gene sequence MT-TS1 (mitochondrially encoded tRNA serine 1) (eg, nonsyndromic hearing loss), full gene sequence NDP (Norrie disease [pseudoglioma]) (eg, Norrie disease), duplication/deletion analysis NHLRC1 (NHL repeat containing 1) (eg, progressive myoclonus epilepsy), full gene sequence PHOX2B (paired-like homeobox 2b) (eg, congenital central hypoventilation syndrome), duplication/deletion analysis PLN (phospholamban) (eg, dilated cardiomyopathy, hypertrophic cardiomyopathy), full gene sequence RHD (Rh blood group, D antigen) (eg, hemolytic disease of the fetus and newborn, Rh maternal/fetal compatibility), deletion analysis (eg, exons 4, 5, and 7, pseudogene) RHD (Rh blood group, D antigen) (eg, hemolytic disease of the fetus and newborn, Rh maternal/fetal compatibility), deletion analysis (eg, exons 4, 5, and 7, pseudogene), performed on cell-free fetal DNA in maternal blood (For human erythrocyte gene analysis of RHD, use a separate unit of 81403) SH2D1A (SH2 domain containing 1A) (eg, X-linked lymphoproliferative syndrome), duplication/deletion analysis TWIST1 (twist homolog 1 [Drosophila]) (eg, Saethre-Chotzen syndrome), duplication/deletion analysis UBA1 (ubiquitin-like modifier activating enzyme 1) (eg, spinal muscular atrophy, X-linked), targeted sequence analysis (eg, exon 15) VHL (von Hippel-Lindau tumor suppressor) (eg, von Hippel-Lindau familial cancer syndrome), deletion/duplication analysis VWF (von Willebrand factor) (eg, von Willebrand disease types 2A, 2B, 2M), targeted sequence analysis (eg, exon 28)
81404Molecular pathology procedure, Level 5 (eg, analysis of 2-5 exons by DNA sequence analysis, mutation scanning or duplication/deletion variants of 6-10 exons, or characterization of a dynamic mutation disorder/triplet repeat by Southern blot analysis) ACADS (acyl-CoA dehydrogenase, C-2 to C-3 short chain) (eg, short chain acyl-CoA dehydrogenase deficiency), targeted sequence analysis (eg, exons 5 and 6) AQP2 (aquaporin 2 [collecting duct]) (eg, nephrogenic diabetes insipidus), full gene sequence ARX (aristaless related homeobox) (eg, X-linked lissencephaly with ambiguous genitalia, X-linked mental retardation), full gene sequence AVPR2 (arginine vasopressin receptor 2) (eg, nephrogenic diabetes insipidus), full gene sequence BBS10 (Bardet-Biedl syndrome 10) (eg, Bardet-Biedl syndrome), full gene sequence BTD (biotinidase) (eg, biotinidase deficiency), full gene sequence C10orf2 (chromosome 10 open reading frame 2) (eg, mitochondrial DNA depletion syndrome), full gene sequence CAV3 (caveolin 3) (eg, CAV3-related distal myopathy, limb-girdle muscular dystrophy type 1C), full gene sequence CD40LG (CD40 ligand) (eg, X-linked hyper IgM syndrome), full gene sequence CDKN2A (cyclin-dependent kinase inhibitor 2A) (eg, CDKN2A-related cutaneous malignant melanoma, familial atypical mole-malignant melanoma syndrome), full gene sequence CLRN1 (clarin 1) (eg, Usher syndrome, type 3), full gene sequence COX6B1 (cytochrome c oxidase subunit VIb polypeptide 1) (eg, mitochondrial respiratory chain complex IV deficiency), full gene sequence CPT2 (carnitine palmitoyltransferase 2) (eg, carnitine palmitoyltransferase II deficiency), full gene sequence CRX (cone-rod homeobox) (eg, cone-rod dystrophy 2, Leber congenital amaurosis), full gene sequence CYP1B1 (cytochrome P450, family 1, subfamily B, polypeptide 1) (eg, primary congenital glaucoma), full gene sequence EGR2 (early growth response 2) (eg, Charcot-Marie-Tooth), full gene sequence EMD (emerin) (eg, Emery-Dreifuss muscular dystrophy), duplication/deletion analysis EPM2A (epilepsy, progressive myoclonus type 2A, Lafora disease [laforin]) (eg, progressive myoclonus epilepsy), full gene sequence FGF23 (fibroblast growth factor 23) (eg, hypophosphatemic rickets), full gene sequence FGFR2 (fibroblast growth factor receptor 2) (eg, craniosynostosis, Apert syndrome, Crouzon syndrome), targeted sequence analysis (eg, exons 8, 10) FGFR3 (fibroblast growth factor receptor 3) (eg, achondroplasia, hypochondroplasia), targeted sequence analysis (eg, exons 8, 11, 12, 13) FHL1 (four and a half LIM domains 1) (eg, Emery-Dreifuss muscular dystrophy), full gene sequence FKRP (fukutin related protein) (eg, congenital muscular dystrophy type 1C [MDC1C], limb-girdle muscular dystrophy [LGMD] type 2I), full gene sequence FOXG1 (forkhead box G1) (eg, Rett syndrome), full gene sequence FSHMD1A (facioscapulohumeral muscular dystrophy 1A) (eg, facioscapulohumeral muscular dystrophy), evaluation to detect abnormal (eg, deleted) alleles FSHMD1A (facioscapulohumeral muscular dystrophy 1A) (eg, facioscapulohumeral muscular dystrophy), characterization of haplotype(s) (ie, chromosome 4A and 4B haplotypes) GH1 (growth hormone 1) (eg, growth hormone deficiency), full gene sequence GP1BB (glycoprotein Ib [platelet], beta polypeptide) (eg, Bernard-Soulier syndrome type B), full gene sequence (For common deletion variants of alpha globin 1 and alpha globin 2 genes, use 81257) HNF1B (HNF1 homeobox B) (eg, maturity-onset diabetes of the young [MODY]), duplication/deletion analysis HRAS (v-Ha-ras Harvey rat sarcoma viral oncogene homolog) (eg, Costello syndrome), full gene sequence HSD3B2 (hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 2) (eg, 3-beta-hydroxysteroid dehydrogenase type II deficiency), full gene sequence HSD11B2 (hydroxysteroid [11-beta] dehydrogenase 2) (eg, mineralocorticoid excess syndrome), full gene sequence HSPB1 (heat shock 27kDa protein 1) (eg, Charcot-Marie-Tooth disease), full gene sequence INS (insulin) (eg, diabetes mellitus), full gene sequence KCNJ1 (potassium inwardly-rectifying channel, subfamily J, member 1) (eg, Bartter syndrome), full gene sequence KCNJ10 (potassium inwardly-rectifying channel, subfamily J, member 10) (eg, SeSAME syndrome, EAST syndrome, sensorineural hearing loss), full gene sequence LITAF (lipopolysaccharide-induced TNF factor) (eg, Charcot-Marie-Tooth), full gene sequence MEFV (Mediterranean fever) (eg, familial Mediterranean fever), full gene sequence MEN1 (multiple endocrine neoplasia I) (eg, multiple endocrine neoplasia type 1, Wermer syndrome), duplication/deletion analysis MMACHC (methylmalonic aciduria [cobalamin deficiency] cblC type, with homocystinuria) (eg, methylmalonic acidemia and homocystinuria), full gene sequence MPV17 (MpV17 mitochondrial inner membrane protein) (eg, mitochondrial DNA depletion syndrome), duplication/deletion analysis NDP (Norrie disease [pseudoglioma]) (eg, Norrie disease), full gene sequence NDUFA1 (NADH dehydrogenase [ubiquinone] 1 alpha subcomplex, 1, 7.5kDa) (eg, Leigh syndrome, mitochondrial complex I deficiency), full gene sequence NDUFAF2 (NADH dehydrogenase [ubiquinone] 1 alpha subcomplex, assembly factor 2) (eg, Leigh syndrome, mitochondrial complex I deficiency), full gene sequence NDUFS4 (NADH dehydrogenase [ubiquinone] Fe-S protein 4, 18kDa [NADH-coenzyme Q reductase]) (eg, Leigh syndrome, mitochondrial complex I deficiency), full gene sequence NIPA1 (non-imprinted in Prader-Willi/Angelman syndrome 1) (eg, spastic paraplegia), full gene sequence NLGN4X (neuroligin 4, X-linked) (eg, autism spectrum disorders), duplication/deletion analysis NPC2 (Niemann-Pick disease, type C2 [epididymal secretory protein E1]) (eg, Niemann-Pick disease type C2), full gene sequence NR0B1 (nuclear receptor subfamily 0, group B, member 1) (eg, congenital adrenal hypoplasia), full gene sequence PDX1 (pancreatic and duodenal homeobox 1) (eg, maturity-onset diabetes of the young [MODY]), full gene sequence PHOX2B (paired-like homeobox 2b) (eg, congenital central hypoventilation syndrome), full gene sequence PLP1 (proteolipid protein 1) (eg, Pelizaeus-Merzbacher disease, spastic paraplegia), duplication/deletion analysis PQBP1 (polyglutamine binding protein 1) (eg, Renpenning syndrome), duplication/deletion analysis PRNP (prion protein) (eg, genetic prion disease), full gene sequence PROP1 (PROP paired-like homeobox 1) (eg, combined pituitary hormone deficiency), full gene sequence PRPH2 (peripherin 2 [retinal degeneration, slow]) (eg, retinitis pigmentosa), full gene sequence PRSS1 (protease, serine, 1 [trypsin 1]) (eg, hereditary pancreatitis), full gene sequence RAF1 (v-raf-1 murine leukemia viral oncogene homolog 1) (eg, LEOPARD syndrome), targeted sequence analysis (eg, exons 7, 12, 14, 17) RET (ret proto-oncogene) (eg, multiple endocrine neoplasia, type 2B and familial medullary thyroid carcinoma), common variants (eg, M918T, 2647_2648delinsTT, A883F) RHO (rhodopsin) (eg, retinitis pigmentosa), full gene sequence RP1 (retinitis pigmentosa 1) (eg, retinitis pigmentosa), full gene sequence SCN1B (sodium channel, voltage-gated, type I, beta) (eg, Brugada syndrome), full gene sequence SCO2 (SCO cytochrome oxidase deficient homolog 2 [SCO1L]) (eg, mitochondrial respiratory chain complex IV deficiency), full gene sequence SDHC (succinate dehydrogenase complex, subunit C, integral membrane protein, 15kDa) (eg, hereditary paraganglioma-pheochromocytoma syndrome), duplication/deletion analysis SDHD (succinate dehydrogenase complex, subunit D, integral membrane protein) (eg, hereditary paraganglioma), full gene sequence SGCG (sarcoglycan, gamma [35kDa dystrophin-associated glycoprotein]) (eg, limb-girdle muscular dystrophy), duplication/deletion analysis SH2D1A (SH2 domain containing 1A) (eg, X-linked lymphoproliferative syndrome), full gene sequence SLC16A2 (solute carrier family 16, member 2 [thyroid hormone transporter]) (eg, specific thyroid hormone
81405Molecular pathology procedure, Level 6 (eg, analysis of 6-10 exons by DNA sequence analysis, mutation scanning or duplication/deletion variants of 11-25 exons, regionally targeted cytogenomic array analysis) Cytogenomic constitutional targeted microarray analysis of chromosome 22q13 by interrogation of genomic regions for copy number and single nucleotide polymorphism (SNP) variants for chromosomal abnormalities (When performing cytogenomic [genome-wide] analysis, for constitutional chromosomal abnormalities. See 81228, 81229, 81349)
81406Molecular pathology procedure, Level 7 (eg, analysis of 11-25 exons by DNA sequence analysis, mutation scanning or duplication/deletion variants of 26-50 exons) ACADVL (acyl-CoA dehydrogenase, very long chain) (eg, very long chain acyl-coenzyme A dehydrogenase deficiency), full gene sequence ACTN4 (actinin, alpha 4) (eg, focal segmental glomerulosclerosis), full gene sequence AFG3L2 (AFG3 ATPase family gene 3-like 2 [S. cerevisiae]) (eg, spinocerebellar ataxia), full gene sequence AIRE (autoimmune regulator) (eg, autoimmune polyendocrinopathy syndrome type 1), full gene sequence ALDH7A1 (aldehyde dehydrogenase 7 family, member A1) (eg, pyridoxine-dependent epilepsy), full gene sequence ANO5 (anoctamin 5) (eg, limb-girdle muscular dystrophy), full gene sequence ANOS1 (anosmin-1) (eg, Kallmann syndrome 1), full gene sequence APP (amyloid beta [A4] precursor protein) (eg, Alzheimer disease), full gene sequence ASS1 (argininosuccinate synthase 1) (eg, citrullinemia type I), full gene sequence ATL1 (atlastin GTPase 1) (eg, spastic paraplegia), full gene sequence ATP1A2 (ATPase, Na+/K+ transporting, alpha 2 polypeptide) (eg, familial hemiplegic migraine), full gene sequence ATP7B (ATPase, Cu++ transporting, beta polypeptide) (eg, Wilson disease), full gene sequence BBS1 (Bardet-Biedl syndrome 1) (eg, Bardet-Biedl syndrome), full gene sequence BBS2 (Bardet-Biedl syndrome 2) (eg, Bardet-Biedl syndrome), full gene sequence BCKDHB (branched-chain keto acid dehydrogenase E1, beta polypeptide) (eg, maple syrup urine disease, type 1B), full gene sequence BEST1 (bestrophin 1) (eg, vitelliform macular dystrophy), full gene sequence BMPR2 (bone morphogenetic protein receptor, type II [serine/threonine kinase]) (eg, heritable pulmonary arterial hypertension), full gene sequence BRAF (B-Raf proto-oncogene, serine/threonine kinase) (eg, Noonan syndrome), full gene sequence BSCL2 (Berardinelli-Seip congenital lipodystrophy 2 [seipin]) (eg, Berardinelli-Seip congenital lipodystrophy), full gene sequence BTK (Bruton agammaglobulinemia tyrosine kinase) (eg, X-linked agammaglobulinemia), full gene sequence CACNB2 (calcium channel, voltage-dependent, beta 2 subunit) (eg, Brugada syndrome), full gene sequence CAPN3 (calpain 3) (eg, limb-girdle muscular dystrophy [LGMD] type 2A, calpainopathy), full gene sequence CBS (cystathionine-beta-synthase) (eg, homocystinuria, cystathionine beta-synthase deficiency), full gene sequence CDH1 (cadherin 1, type 1, E-cadherin [epithelial]) (eg, hereditary diffuse gastric cancer), full gene sequence CDKL5 (cyclin-dependent kinase-like 5) (eg, early infantile epileptic encephalopathy), full gene sequence CLCN1 (chloride channel 1, skeletal muscle) (eg, myotonia congenita), full gene sequence CLCNKB (chloride channel, voltage-sensitive Kb) (eg, Bartter syndrome 3 and 4b), full gene sequence CNTNAP2 (contactin-associated protein-like 2) (eg, Pitt-Hopkins-like syndrome 1), full gene sequence COL6A2 (collagen, type VI, alpha 2) (eg, collagen type VI-related disorders), duplication/deletion analysis CPT1A (carnitine palmitoyltransferase 1A [liver]) (eg, carnitine palmitoyltransferase 1A [CPT1A] deficiency), full gene sequence CRB1 (crumbs homolog 1 [Drosophila]) (eg, Leber congenital amaurosis), full gene sequence CREBBP (CREB binding protein) (eg, Rubinstein-Taybi syndrome), duplication/deletion analysis DBT (dihydrolipoamide branched chain transacylase E2) (eg, maple syrup urine disease, type 2), full gene sequence DLAT (dihydrolipoamide S-acetyltransferase) (eg, pyruvate dehydrogenase E2 deficiency), full gene sequence DLD (dihydrolipoamide dehydrogenase) (eg, maple syrup urine disease, type III), full gene sequence DSC2 (desmocollin) (eg, arrhythmogenic right ventricular dysplasia/cardiomyopathy 11), full gene sequence DSG2 (desmoglein 2) (eg, arrhythmogenic right ventricular dysplasia/cardiomyopathy 10), full gene sequence DSP (desmoplakin) (eg, arrhythmogenic right ventricular dysplasia/cardiomyopathy 8), full gene sequence EFHC1 (EF-hand domain [C-terminal] containing 1) (eg, juvenile myoclonic epilepsy), full gene sequence EIF2B3 (eukaryotic translation initiation factor 2B, subunit 3 gamma, 58kDa) (eg, leukoencephalopathy with vanishing white matter), full gene sequence EIF2B4 (eukaryotic translation initiation factor 2B, subunit 4 delta, 67kDa) (eg, leukoencephalopathy with vanishing white matter), full gene sequence EIF2B5 (eukaryotic translation initiation factor 2B, subunit 5 epsilon, 82kDa) (eg, childhood ataxia with central nervous system hypomyelination/vanishing white matter), full gene sequence ENG (endoglin) (eg, hereditary hemorrhagic telangiectasia, type 1), full gene sequence EYA1 (eyes absent homolog 1 [Drosophila]) (eg, branchio-oto-renal [BOR] spectrum disorders), full gene sequence F8 (coagulation factor VIII) (eg, hemophilia A), duplication/deletion analysis FAH (fumarylacetoacetate hydrolase [fumarylacetoacetase]) (eg, tyrosinemia, type 1), full gene sequence FASTKD2 (FAST kinase domains 2) (eg, mitochondrial respiratory chain complex IV deficiency), full gene sequence FIG4 (FIG4 homolog, SAC1 lipid phosphatase domain containing [S. cerevisiae]) (eg, Charcot-Marie-Tooth disease), full gene sequence FTSJ1 (FtsJ RNA methyltransferase homolog 1 [E. coli]) (eg, X-linked mental retardation 9), full gene sequence FUS (fused in sarcoma) (eg, amyotrophic lateral sclerosis), full gene sequence GAA (glucosidase, alpha; acid) (eg, glycogen storage disease type II [Pompe disease]), full gene sequence GALC (galactosylceramidase) (eg, Krabbe disease), full gene sequence GALT (galactose-1-phosphate uridylyltransferase) (eg, galactosemia), full gene sequence GARS (glycyl-tRNA synthetase) (eg, Charcot-Marie-Tooth disease), full gene sequence GCDH (glutaryl-CoA dehydrogenase) (eg, glutaricacidemia type 1), full gene sequence GCK (glucokinase [hexokinase 4]) (eg, maturity-onset diabetes of the young [MODY]), full gene sequence GLUD1 (glutamate dehydrogenase 1) (eg, familial hyperinsulinism), full gene sequence GNE (glucosamine [UDP-N-acetyl]-2-epimerase/N-acetylmannosamine kinase) (eg, inclusion body myopathy 2 [IBM2], Nonaka myopathy), full gene sequence GRN (granulin) (eg, frontotemporal dementia), full gene sequence HADHA (hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA hydratase [trifunctional protein] alpha subunit) (eg, long chain acyl-coenzyme A dehydrogenase deficiency), full gene sequence HADHB (hydroxyacyl-CoA dehydrogenase/3-ketoacyl-CoA thiolase/enoyl-CoA hydratase [trifunctional protein], beta subunit) (eg, trifunctional protein deficiency), full gene sequence HEXA (hexosaminidase A, alpha polypeptide) (eg, Tay-Sachs disease), full gene sequence HLCS (HLCS holocarboxylase synthetase) (eg, holocarboxylase synthetase deficiency), full gene sequence HMBS (hydroxymethylbilane synthase) (eg, acute intermittent porphyria), full gene sequence HNF4A (hepatocyte nuclear factor 4, alpha) (eg, maturity-onset diabetes of the young [MODY]), full gene sequence IDUA (iduronidase, alpha-L-) (eg, mucopolysaccharidosis type I), full gene sequence INF2 (inverted formin, FH2 and WH2 domain containing) (eg, focal segmental glomerulosclerosis), full gene sequence IVD (isovaleryl-CoA dehydrogenase) (eg, isovaleric acidemia), full gene sequence JAG1 (jagged 1) (eg, Alagille syndrome), duplication/deletion analysis JUP (junction plakoglobin) (eg, arrhythmogenic right ventricular dysplasia/cardiomyopathy 11), full gene sequence KCNH2 (potassium voltage-gated channel, subfamily H [eag-related], member 2) (eg, short QT syndrome, long QT syndrome), full gene sequence KCNQ1 (potassium voltage-gated channel, KQT-like subfamily, member 1) (eg, short QT syndrome, long QT syndrome), full gene sequence KCNQ2 (potassium voltage-gated channel, KQT-like subfamily, member 2) (eg, epileptic encephalopathy), full gene sequence LDB3 (LIM domain binding 3) (eg, familial dilated cardiomyopathy, myofibrillar myopathy), full gene sequence LDLR (low den
81407Molecular pathology procedure, Level 8 (eg, analysis of 26 50 exons by DNA sequence analysis, mutation scanning or duplication/deletion variants of &gt;50 exons, sequence analysis of multiple genes on one platform)
81419Epilepsy genomic sequence analysis panel, must include analyses for ALDH7A1, CACNA1A, CDKL5, CHD2, GABRG2, GRIN2A, KCNQ2, MECP2, PCDH19, POLG, PRRT2, SCN1A, SCN1B, SCN2A, SCN8A, SLC2A1, SLC9A6, STXBP1, SYNGAP1, TCF4, TPP1, TSC1, TSC2, and ZEB2

References: Alsubaie L, Aloraini T, Amoudi M, et al.(2020) Genomic testing and counseling: The contribution of next-generation sequencing to epilepsy genetics. Ann Hum Genet. Nov 2020; 84(6): 431-436. PMID 32533790

Bagnall RD, Crompton DE, Petrovski S, et al.(2016) Exome-based analysis of cardiac arrhythmia, respiratory control, and epilepsy genes in sudden unexpected death in epilepsy. Ann Neurol. Apr 2016;79(4):522-534. PMID 26704558

Berg AT, Berkovic SF, Brodie MJ et al.(2010) Revised terminology and concepts for organization of seizures and epilepsies: report of the ILAE Commission on Classification and Terminology, 2005-2009. Epilepsia 2010; 51(4):676-85.

Berg AT, Coryell J, Saneto RP, et al.(2017) Early-life epilepsies and the emerging role of genetic testing. JAMA Pediatr. Sep 1 2017;171(9):863-871. PMID 28759667

Butler KM, da Silva C, Alexander JJ, et al.(2017) Diagnostic Yield From 339 Epilepsy Patients Screened on a Clinical Gene Panel. Pediatr Neurol. Dec 2017;77:61-66. PMID 29056246

Cavalleri GL, McCormack M, Alhusaini S et al.(2011) Pharmacogenomics and epilepsy: the road ahead. Pharmacogenomics 2011; 12(10):1429-47.

Chambers C, Jansen LA, Dhamija R.(2016) Review of commercially available epilepsy genetic panels. J Genet Couns. Apr 2016;25(2):213-217. PMID 26536886

Coll M, Allegue C, Partemi S, et al.(2016) Genetic investigation of sudden unexpected death in epilepsy cohort by panel target resequencing. Int J Legal Med. Mar 2016;130(2):331-339. PMID 26423924

Cordoba M, Consalvo D, Moron DG et al.(2012) SLC6A4 gene variants and temporal lobe epilepsy susceptibility: a meta-analysis. Mol Biol Rep 2012; 39(12):10615-9.

Deprez L, Jansen A, De Jonghe P.(2009) Genetics of epilepsy syndromes starting in the first year of life. Neurology. Jan 20 2009; 72(3): 273-81. PMID 19153375

Epicure Consortium, Consortium EM, Steffens M et al.(2012) Genome-wide association analysis of genetic generalized epilepsies implicates susceptibility loci at 1q43, 2p16.1, 2q22.3 and 17q21.32. Hum Mol Genet 2012; 21(24):5359-72.

Epilepsies: diagnosis and management. Clinical guideline 2012; CG137:https://www.nice.org.uk/guidance/cg137/resources/epilepsies-diagnosis-and-management- 35109515407813. Accessed January 26, 2016.

Esterhuizen AI, Mefford HC, Ramesar RS, et al.(2018) Dravet syndrome in South African infants: Tools for an early diagnosis. Seizure. Nov 2018;62:99-105. PMID 30321769

Fisher RS, Cross JH, French JA, et al.(2017) Operational classification of seizure types by the International League Against Epilepsy: Position Paper of the ILAE Commission for Classification and Terminology. Epilepsia. Apr 2017; 58(4): 522-530. PMID 28276060

Food and Drug Administration (FDA).(2018) Label: Tegretol. 2018; https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/016608s115_018281_s058_018927s055_020234_s047.pdf. Accessed January 8, 2021.

GeneTests. https://www.genetests.org/. Accessed January 5, 2017.

Go CY, Mackay MT, Weiss SK, et al.(2012) Evidence-based guideline update: medical treatment of infantile spasms. Report of the Guideline Development Subcommittee of the American Academy of Neurology and the Practice Committee of the Child Neurology Society. Neurology. Jun 12 2012;78(24):1974-1980. PMID 22689735

Guo Y, Baum LW, Sham PC et al(2012) Two-stage genome-wide association study identifies variants in CAMSAP1L1 as susceptibility loci for epilepsy in Chinese. Hum Mol Genet 2012; 21(5):1184-9.

Guo Y, Yan KP, Qu Q, et al.(2015) Common variants of KCNJ10 are associated with susceptibility and anti-epileptic drug resistance in Chinese genetic generalized epilepsies. PLoS One. 2015;10(4):e0124896. PMID 25874548

Haerian BS, Roslan H, Raymond AA et al.(2010) ABCB1 C3435T polymorphism and the risk of resistance to antiepileptic drugs in epilepsy: a systematic review and meta-analysis. Seizure 2010; 19(6):339-46.

Heinzen EL, Depondt C, Cavalleri GL et al.(2012) Exome sequencing followed by large-scale genotyping fails to identify single rare variants of large effect in idiopathic generalized epilepsy. Am J Hum Genet 2012; 91(2):293-302.

Helbig I, Lowenstein DH.(2013) Genetics of the epilepsies: where are we and where are we going? Curr Opin Neurol 2013; 26(2):179-85.

Hesse AN, Bevilacqua J, Shankar K, et al.(2018) Retrospective genotype-phenotype analysis in a 305 patient cohort referred for testing of a targeted epilepsy panel. Epilepsy Res. Aug 2018;144:53-61. PMID 29778030

Hirose S, Scheffer IE, Marini C et al.(2013) SCN1A testing for epilepsy: application in clinical practice. Epilepsia 2013; 54(5):946-52.

Hoelz H, Herdl C, Gerstl L, et al.(2020) Impact on Clinical Decision Making of Next-Generation Sequencing in Pediatric Epilepsy in a Tertiary Epilepsy Referral Center. Clin EEG Neurosci. Jan 2020; 51(1): 61-69. PMID 31554424

Jang SY, Kim MK, Lee KR et al.(2009) Gene-to-gene interaction between sodium channel-related genes in determining the risk of antiepileptic drug resistance. J Korean Med Sci 2009; 24(1):62-8.

Jiang T, Gao J, Jiang L, et al.(2021) Application of Trio-Whole Exome Sequencing in Genetic Diagnosis and Therapy in Chinese Children With Epilepsy. Front Mol Neurosci. 2021; 14: 699574. PMID 34489640

Johannesen KM, Nikanorova N, Marjanovic D, et al.(2020) Utility of genetic testing for therapeutic decision-making in adults with epilepsy. Epilepsia. Jun 2020; 61(6): 1234-1239. PMID 32427350

Kasperaviciute D, Catarino CB, Heinzen EL et al.(2010) Common genetic variation and susceptibility to partial epilepsies: a genome-wide association study. Brain 2010; 133(Pt 7):2136-47.

Kauffman MA, Moron DG, Consalvo D et al.(2008) Association study between interleukin 1 beta gene and epileptic disorders: a HuGe review and meta-analysis. Genet Med 2008; 10(2):83-8.

Kim SY, Jang SS, Kim H, et al.(2021) Genetic diagnosis of infantile-onset epilepsy in the clinic: Application of whole-exome sequencing following epilepsy gene panel testing. Clin Genet. Mar 2021; 99(3): 418-424. PMID 33349918

Kwan P, Brodie MJ.(2000) Early identification of refractory epilepsy, N Engl J Med 2000; 342(5):314-9.

Kwan P, Poon WS, Ng HK et al.(2008) Multidrug resistance in epilepsy and polymorphisms in the voltage-gated sodium channel genes SCN1A, SCN2A, and SCN3A: correlation among phenotype, genotype, and mRNA expression. Pharmacogenet Genomics 2008; 18(11):989-98.

Lin CH, Chou IC, Hong SY.(2021) Genetic factors and the risk of drug-resistant epilepsy in young children with epilepsy and neurodevelopment disability: A prospective study and updated meta-analysis. Medicine (Baltimore). Mar 26 2021; 100(12): e25277. PMID 33761731

Lindy AS, Stosser MB, Butler E, et al.(2018) Diagnostic outcomes for genetic testing of 70 genes in 8565 patients with epilepsy and neurodevelopmental disorders. Epilepsia. May 2018;59(5):1062-1071. PMID 29655203

Lu Y, Fang Y, Wu X, et al.(2016) Effects of UGT1A9 genetic polymorphisms on monohydroxylated derivative of oxcarbazepine concentrations and oxcarbazepine monotherapeutic efficacy in Chinese patients with epilepsy. Eur J Clin Pharmacol. Nov 29 2016. PMID 27900402

McKnight D, Bristow SL, Truty RM, et al.(2022) Multigene Panel Testing in a Large Cohort of Adults With Epilepsy: Diagnostic Yield and Clinically Actionable Genetic Findings. Neurol Genet. Feb 2022; 8(1): e650. PMID 34926809

Merwick A, O'Brien M, Delanty N.(2012) Complex single gene disorders and epilepsy. Epilepsia 2012; 53 Suppl 4:81-91.

Miao P, Feng J, Guo Y, et al.(2018) Genotype and phenotype analysis using an epilepsy-associated gene panel in Chinese pediatric epilepsy patients. Clin Genet. Dec 2018;94(6):512-520. PMID 30182498

Miller IO, Sotero de Menezes MA.(2014) SCN1A-Related Seizure Disorders. In: Adam MP, Ardinger HH, Pagon RA, et al., eds. GeneReviews. Seattle, WA: University of Washington; 2014.

Minardi R, Licchetta L, Baroni MC, et al.(2020) Whole-exome sequencing in adult patients with developmental and epileptic encephalopathy: It is never too late. Clin Genet. Nov 2020; 98(5): 477-485. PMID 32725632

Moller RS, Larsen LH, Johannesen KM, et al.(2016) Gene panel testing in epileptic encephalopathies and familial epilepsies. Mol Syndromol. Sep 2016;7(4):210-219. PMID 27781031

Mulley JC, Nelson P, Guerrero S et al.(2006) A new molecular mechanism for severe myoclonic epilepsy of infancy: exonic deletions in SCN1A. Neurology 2006; 67(6):1094-5.

National Center for Biotechnology Information,(2017) Genetic Testing Registry. https://www.ncbi.nlm.nih.gov/gtr/. Accessed January 5, 2017.

National Institute of Neurological Disorders and Stroke (NINDS).(2017) NINDS Common Data Elements. Epilepsy. 2017, January 31; https://www.commondataelements.ninds.nih.gov/Epilepsy.aspx#tab=Data_Standards. Accessed February 7, 2017.

Nurmohamed L, Garcia-Bournissen F, Buono RJ et al.(2010) Predisposition to epilepsy--does the ABCB1 gene play a role? Epilepsia 2010; 51(9):1882-5.

Ottman R, Hirose S, Jain S et al.(2010) Genetic testing in the epilepsies--report of the ILAE Genetics Commission. Epilepsia 2010; 51(4):655-70.

Palmer EE, Sachdev R, Macintosh R, et al.(2021) Diagnostic Yield of Whole Genome Sequencing After Nondiagnostic Exome Sequencing or Gene Panel in Developmental and Epileptic Encephalopathies. Neurology. Mar 30 2021; 96(13): e1770-e1782. PMID 33568551

Pellock JM, Hrachovy R, Shinnar S, et al.(2010) Infantile spasms: a U.S. consensus report. Epilepsia. Oct 2010;51(10):2175-2189. PMID 20608959

Peng J, Pang N, Wang Y, et al.(2019) Next-generation sequencing improves treatment efficacy and reduces hospitalization in children with drug-resistant epilepsy. CNS Neurosci Ther. Jan 2019;25(1):14-20. PMID 29933521

Petrovski S, Kwan P.(2013) Unraveling the genetics of common epilepsies: approaches, platforms, and caveats. Epilepsy Behav 2013; 26(3):229-33.

Riviello JJ, Jr., Ashwal S, Hirtz D, et al.(2006) Practice parameter: diagnostic assessment of the child with status epilepticus (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology and the Practice Committee of the Child Neurology Society. Neurology. Nov 14 2006;67(9):1542-1550. PMID 17101884

Salinas V, Martinez N, Maturo JP, et al.(2021) Clinical next generation sequencing in developmental and epileptic encephalopathies: Diagnostic relevance of data re-analysis and variants re-interpretation. Eur J Med Genet. Dec 2021; 64(12): 104363. PMID 34673242

Scheffer IE, Bennett CA, Gill D, et al.(2023) Exome sequencing for patients with developmental and epileptic encephalopathies in clinical practice. Dev Med Child Neurol. Jan 2023; 65(1): 50-57. PMID 35701389

Societies EFoN.(2010) EFNS Guidelines on the Molecular Diagnosis of Channelopathies, Epilepsies, Migraine, Stroke, and Dementias. 2010; http://onlinelibrary.wiley.com/doi/10.1111/j.1468- 1331.2010.02985.x/pdf. Accessed December 8, 2014.

Sun D, Liu Y, Cai W, et al.(2021) Detection of Disease-Causing SNVs/Indels and CNVs in Single Test Based on Whole Exome Sequencing: A Retrospective Case Study in Epileptic Encephalopathies. Front Pediatr. 2021; 9: 635703. PMID 34055682

Tan NC, Berkovic SF.(2010) The Epilepsy Genetic Association Database (epiGAD): analysis of 165 genetic association studies, 1996-2008. Epilepsia 2010; 51(4):686-9.

Tang L, Lu X, Tao Y, et al.(2014) SCN1A rs3812718 polymorphism and susceptibility to epilepsy with febrile seizures: a meta-analysis. Gene. Jan 1 2014;533(1):26-31. PMID 24076350

Trump N, McTague A, Brittain H, et al.(2016) Improving diagnosis and broadening the phenotypes in early-onset seizure and severe developmental delay disorders through gene panel analysis. J Med Genet. May 2016;53(5):310-317. PMID 26993267

Williams CA, Battaglia A.(2013) Molecular biology of epilepsy genes. Exp Neurol 2013; 244:51-8.

Wilmshurst JM, Gaillard WD, Vinayan KP, et al.(2015) Summary of recommendations for the management of infantile seizures: Task Force Report for the ILAE Commission of Pediatrics. Epilepsia. Aug 2015;56(8):1185-1197. PMID 26122601

Wirrell EC, Laux L, Donner E, et al.(2017) Optimizing the Diagnosis and Management of Dravet Syndrome: Recommendations From a North American Consensus Panel. Pediatr Neurol. Mar 2017; 68: 18-34.e3. PMID 28284397


Group specific policy will supersede this policy when applicable. This policy does not apply to the Wal-Mart Associates Group Health Plan participants or to the Tyson Group Health Plan participants.
CPT Codes Copyright © 2024 American Medical Association.