Abstract
Genetic diseases test the functionality of an infant’s genome during fetal-neonatal adaptation and represent a leading cause of neonatal and infant mortality in the US. Due to disease acuity, gene locus and allelic heterogeneity, and overlapping/diverse clinical phenotypes, diagnostic genome sequencing in neonatal intensive care units has required development of methods to shorten turnaround time and to improve genomic interpretation. Between 2012 and 2021, 33 clinical studies have documented diagnostic and clinical utility of first-tier, rapid or ultra-rapid whole genome sequencing through cost-effective identification of pathogenic genomic variants which change medical management, suggest new therapeutic strategies, and refine prognosis. Genomic diagnosis also permits prediction of reproductive recurrence risk for parents and surviving probands. Using implementation science and quality improvement, deployment of a Genomic Learning Healthcare System will contribute to reduction of neonatal and infant mortality through integration of genome sequencing into best practice neonatal intensive care.
Keywords: Genomics, Neonatal Intensive Care Unit, Newborn infants, Precision medicine
Introduction: the unique genomics of neonatology
The perinatal period, defined as six months before to 12 months after birth, is unique in terms of healthcare consequences across the lifetime. During this period, the functionality of a child’s genome is tested by the biologic requirements of fetal-neonatal transition and extra-uterine physiologic adaptation. During pregnancy and the immediate post-partum period, many fetal physiologic processes are provided, or compensated for, by the maternal or placental genome. In addition, genome-encoded developmental regulation continues to activate and silence genes and gene pathways throughout the first months and years of life. For example, drug metabolism is quite different in newborns and older children due to lack of expression of the key metabolic enzyme cytochrome P450 2D6 (CYP2D6) (92). Finally, delivery of viable fetuses may occur at diverse, post-conception time points. Thus, in caring for newborns, neonatologists must consider age both as days since conception and days since delivery. Much remains to be learned about the factors that regulate the ontogeny of gene expression and silencing associated with neonatal and gestational ages (92). Thus, genetic, genomic, and functional genomic regulation of fetal development and neonatal adaptation to the extra-uterine environment is uniquely challenging in three ways. Firstly, the timing of onset of monogenic disease is regulated by the continuum of causal gene expression in the fetus or newborn during the perinatal period as suggested by examples of genetic disruption of liver and cerebral function (68, 88). Secondly, the timing of onset of monogenic disease is influenced by the continuum of compensatory effects of maternal or placental genes. For example, in the setting of primary neonatal immunodeficiency, maternal immunoglobulins may protect a newborn from infections for months (19, 95). Thirdly, the adaptive physiologic changes that occur at delivery immediately expose latent genetic defects. Third trimester echocardiography in fetuses with congenital heart disease, for example, can define cardiac anatomy but may not be able to predict neonatal cardiac adaptation to extra-uterine life.
While we can now routinely decode and analyze a human genome in a day, the contributions of many genetic defects to disease penetrance, severity, onset, complications, response to treatment, and outcomes need to be defined to permit clinically actionable genomic results and remain under active investigation (40)(75). Similar to comparing a perfect space rocket at t minus 5 minutes for launch and t plus five minutes, the phenotypically normal fetus may develop life-threatening genetic disease in the neonatal period whose treatment and prognosis may be difficult to predict based on fetal phenotype. For example, knowledge of the underlying genetics in developmental epileptic encephalopathies does not necessarily translate today into clinically actionable guidance for treatment or prognosis. While we have identified genetic variants associated with >7,000 genetic diseases, for a majority, we do not yet understand the encoded mechanisms of action. Another challenge to use of genomic information for sick infants is that different variants in a single gene may be associated with different disease phenotypes (76), and similar disease phenotypes can be associated with different genes (55). The former is particularly perplexing. Variants in a voltage-gated potassium channel gene, KCNQ2, for example, map to both Developmental and epileptic encephalopathy type 7 (DEE7, Mendelian Inheritance in Man (MIM) #613720) and Benign neonatal seizures type 1 (BNS1, MIM#12100, also known as Myokimia). The treatment and prognosis of these two disorders are quite different. BNS1 presents with seizure onset at 2-8 days of life, seizures respond to first-line antiepileptic medications, such as phenobarbital, and most remit by 6 weeks. DEE7 has similar age of seizure onset but requires much more aggressive antiepileptic therapy and is associated with developmental delay and intellectual disability. In summary, while the greatest potential contribution of genome sequencing to human health is currently for newborns, the unique physiologic and genomic characteristics of the newborn plus our incomplete understanding of encoded mechanisms of action, treatment guidance, and prognosis present bottlenecks to achieving the full potential of timely genetic diagnosis for reducing fetal and neonatal morbidity and mortality. However, the importance of the contributions of genetic disease to neonatal and infant mortality has accelerated study and integration of genome sequencing into the neonatal intensive care unit (NICU) to improve infant outcomes.
The value of genome sequencing in the NICU –
Although many of the physiologically supportive clinical practices of neonatal intensive care help to improve survival of critically ill infants with genetic diseases, many affected infants have unique disease mechanisms associated with pathogenic genomic variants that are rare or novel due to reduced reproductive fitness (49) and that disrupt function and/or anatomy of multiple organs (91). An evidence-based clinical practice guideline from the American College of Medical Genetics and Genomics (ACMG) supports the clinical utility and desirable effects of whole exome (WES) and whole genome sequencing (WGS) on active and long-term clinical management for pediatric patients <1 year of age with 1 or more congenital anomalies (56). NICUs have recognized the critical need for diagnostic discovery of genomic causes of newborn diseases to individualize treatment, establish prognosis, predict reproductive recurrence risk, and improve infant outcomes (44, 106). Leveraging collaborations with geneticists, genetic counselors, obstetricians, genomicists, and bioinformaticians, advances in genome sequencing technology (especially reduction in turnaround time (75)), computational strategies for identifying pathogenic variants (61), and reduced sequencing costs, NICUs have begun transitioning from a phenotype first to a genotype first approach for genetic diagnosis in critically ill infants by integrating rapid trio WES and WGS into early diagnostic testing that will permit faster diagnosis, more precise prognosis, and rapid identification of individualized treatment options (43, 75). In addition, genotype first diagnosis provides families estimates of reproductive recurrence risk for future pregnancies and for the surviving proband and siblings. To illustrate the diagnostic and therapeutic value of WES and WGS for NICU patients with congenital anomalies, we will briefly describe an example, epileptic encephalopathy due to thiamine metabolism dysfunction syndrome.
Epileptic encephalopathy due to thiamine metabolism dysfunction syndrome –
Although infections, trauma, and environmental factors are associated with epileptic encephalopathies, genetic etiologies contribute significantly to their frequency and pathogenesis (6, 93). Due to purifying selection, genetic causes are heterogeneous. Discovery of a monogenic etiology in an infant with urWGS can inform precision therapy as illustrated by a recently reported case (75). Briefly, a 5-week-old, previously healthy male infant of consanguineous parents whose reproductive history included a previous child who expired with epileptic encephalopathy presented with epileptic encephalopathy. Ultra-rapid (within 16.5 hours) WGS revealed homozygous, known pathogenic (ClinVar VCV000533549 .2) frameshift variants in SLC19A3, a thiamine transporter known to be a monogenic cause of thiamine metabolism dysfunction syndrome 2 (THMD2) (MIM#607483). Based on these findings, treatment with thiamine and biotin was initiated with almost immediate resolution of seizures. At 7 months of age, the infant is reportedly thriving. Untreated THMD2 leads to rapid neurologic deterioration and death (52). This patient illustrates the importance of integrating rapid- or ultra-rapid-WGS for critically ill infants with congenital anomalies that include epileptic encephalopathies, metabolic disorders, and structural birth defects.
Integrating Genome Sequencing in Neonatal Intensive Care Units
The opportunity for genetics and genomics to reduce neonatal morbidity and mortality has occurred over the last 2 decades after neonatal intensive care units (NICUs) developed life-sustaining obstetric, surgical, and medical strategies for infants born at the edge of viability, with life-limiting congenital anomalies, with neonatal infections, and with severe intrapartum insults. In the early 20th century, newborns were delivered at home and survived if they were able to establish respiration, maintain oral intake, and avoid infection. Between 1930 and 1950, deliveries shifted from homes to hospitals to gain access to maternal anesthesia and analgesia. Hospital staff and families recognized that survival of premature infants could be significantly improved by providing thermoregulation, oxygen, hand hygiene, and more intensive physiologic monitoring. This recognition prompted the establishment of the first NICUs in the 1960s. With increased neonatal specialization and favorable reimbursement, rapid expansion of the number of NICUs occurred over the past 3 decades to about 800 units in the United States (30). Concurrent with broad deployment and refinement of neonatal intensive care, the neonatal mortality rate (death within the first 28 days of life) has fallen from 18.7 (1960) to 3.8 (2018) per 1,000 live births. This survival improvement is attributable to collaboration of multidisciplinary teams of nurses, physicians (neonatologists, obstetricians, anesthesiologists, and pediatric subspecialists), respiratory, occupational, and physical therapists, social workers, pharmacists, and clinical investigators focused on the unique physiologic challenges of extra-uterine adaptation. These teams developed, implemented, and deployed quality improvement frameworks with specific care practices, interventions, and quantitative clinical guidelines specific for the newborn’s unique transitional physiology and disease susceptibilities. For example, interventions to improve pulmonary outcomes for premature infants (e.g., antenatal glucocorticoid administration, surfactant replacement therapy, and less invasive ventilation strategies) combined to reduce a previously common cause of neonatal mortality in preterm infants, respiratory distress syndrome, from 20% (1980) to 2% (2019) of infant deaths. As a consequence, the current, most frequent, population-based causes of infant deaths in the United States based on birth certificate data are genetic, and include congenital malformations, deformations, and chromosomal abnormalities, which accounted for 20.6% of 20,921 total infant deaths in the United States in 2019 (2, 36). Many, but not all, of these are caused by single locus genetic diseases. Approximately 10% of the US annual birth cohort or ~400,000 newborns are now admitted to NICUs, costing at least $26 billion (2007 dollars) and accounting for up to 50% of the total United States pediatric health care expenditure (34). In addition, 10% to 25% of critically ill infants in NICUs are estimated to have an undiagnosed single gene locus disorder which may be missed due to the nonspecific presentations of many genetic diseases in the newborn period and to limitations in reimbursement for comprehensive inpatient genetic testing (35, 58, 105, 106).
Prior to availability of genome sequencing in the NICU, genetic diagnosis in critically ill newborn infants relied on both a prenatal and postnatal phenotype first approach. Strategies to assess fetal phenotype, including ultrasound, maternal biochemical testing, and fetal magnetic resonance imaging, provide fetal characteristics that may not be specific to individual genetic diagnoses (35). Historically, fetal and neonatal genetic testing was highly selective and employed chromosomal analysis (karyotyping, fluorescence in situ hybridization, and chromosomal microarray (CMA)) and Sanger sequencing of exons of specific genes. Samples for fetal DNA extraction included chorionic villi, amniocentesis fluid, fetal umbilical blood, or maternal cell free DNA. Diagnoses were limited to aneuploidy, large copy number variants, and recurrent single locus genetic diseases with pathognomonic features (10, 35, 41, 47, 50, 104). Until very recently, there was considerable uncertainty in estimates of the incidence or types of genetic diseases among infants and children hospitalized for critical illness (54, 59). Furthermore, the etiology of common presentations was frequently regarded as polygenic, influenced heavily by the common disease/common variant hypothesis and results of genome-wide association studies. We now know that infant-onset conditions such as epilepsy, developmental delay, intellectual disability, hearing loss, and inflammatory bowel disease are actually each reflective of at least one thousand separate single locus genetic diseases. In addition to a lack of comprehensive testing modalities, patenting of disease genes created barriers to diagnostic testing (39, 46).
Comprehensive genetic testing first became available with the advent of next generation sequencing in 2005 (57). The first individual human genomes were sequenced at a cost of at least a million dollars during the period from 2007 – 2009 (42, 53). This uniquely exciting time in human genetics generated multiple novel realizations. For example, comprehensive sequencing revealed that genomes contained many more variants than suspected (about 6 million per individual). Disease gene discovery was less computationally challenging when trio genome sequencing (parents plus proband) replaced the traditional approach of linkage analysis and positional cloning. An exciting development in 2009 and 2010 was the use of WES and gene panel exome sequencing (71). The exome refers to the collection of all exons of ~20,000 protein-coding genes and comprises about 2% of the genome. While exomes must be sequenced much more deeply than genomes (90-fold versus 20-fold) for comprehensive coverage (because of coverage skewing), they decreased the cost of genome sequencing about tenfold (46). The era of the $1000 research genome led to many discoveries. Almost overnight, human genetics went from being descriptive to quantitative. Advances in bioinformatics facilitated the development of high performance computing strategies necessary for genomic data management and interpretation. We realized that human genomes had many more de novo variants than previously recognized (about 80 per genome), and that resultant dominant disorders contributed the majority of single locus genetic disease diagnoses in outbred populations (48). The success of positional cloning for identification of recessive conditions led to the erroneous conclusion that they were much more common than dominant disorders. The first use of exome sequencing to diagnose genetic diseases was in 2009 (15, 70), and the first gene panel sequencing test for diagnosis and carrier testing was in 2011 (7). Two landmark papers at that time showed the ability of WGS- or WES-based diagnosis dramatically to change childhood outcomes (4, 107). Many physician-scientists who had trained in adult internal medicine believing it to be the most rigorous clinical discipline (for example, one of us (SFK)) suddenly realized that the next decade would be dominated by pediatric molecular discoveries! We realized to our great chagrin that 27% of variants in our databases had been misclassified as pathogenic (7). As a community, we had been “looking under the lamp-post” for our lost diagnostic keys and thereby over-interpreting pathogenicity.
Rapid, diagnostic WGS of infants with suspected genetic diseases in NICUs became possible in 2012 (83). Until that time, time to result by genome or exome sequencing was several months, limiting diagnostic use to outpatients (Figure 1). Faster sequencing and bioinformatics, together with semi-automated diagnostic interpretation, decreased time to genome sequencing result to 50 hours.
Figure 1.
The current inflection point in precision neonatology for single locus genetic diseases. The cost of research-grade genome sequencing (black line) and time to result of diagnostic rWGS (blue line) have been decreasing for ten years. The number of known genetic diseases (yellow line) has increased dramatically since the advent of next generation sequencing. The number of approved genetic therapies for childhood-onset genetic diseases (teal line) is rapidly increasing.
In the past ten years, the turnaround time, scalability, cost, and diagnostic yield of rapid, diagnostic WGS have continued to improve iteratively (Figure 1). Minimum turnaround time is now 13.5 hours (75). WGS is scalable to entire populations (29). Minimum cost of a research genome is now ~$500 (37). Diagnostic capacity has steadily expanded to include disorders of the mitochondrial genome, structural and copy number variants, simple sequence repeat expansions, imprinting disorders, and loci featuring pseudogenes. Sensitivity and specificity for single nucleotide variants now exceed 99.5% (74). Rapid diagnostic exome sequencing and rapid diagnostic gene panel exome sequencing became available shortly after rapid genome sequencing. They remain less expensive than genome sequencing ($2000-$2500 for rapid, diagnostic gene panels, $4000-$5000 for rapid exome sequencing, and $8,000-$10,000 for rapid genome sequencing). They cannot, however, be performed as rapidly as genome sequencing, because they require exon enrichment and amplification steps. While their diagnostic yield has been similar to that of genome sequencing, as our ability to detect pathogenic, non-exonic variants improves, genome sequencing will inevitably become superior.
Rapid Genome Sequencing Technology
The technology, infrastructure, and computational strategies that enable rapid NICU genome sequencing have become highly standardized (Figure 2). Firstly, parental consent is sought. This consent is necessary because of the possibility of harm for the infant. In the United States, most potential harm was mitigated by the Genetic Information Nondiscrimination Act (GINA) of 2008. The scope of testing must also be determined with parental consent. Parent – infant trio testing is faster and has a higher potential diagnostic yield than singleton testing (17). Some parents, however, such as armed forces service members, are not protected by GINA. Parents must also decide about whether they wish incidental findings to be returned (66, 67). These genomic variants are not related to the infant’s current illness but which have significant consequences for future health (67). Finally, there is the option of rapid and ultra-rapid testing. The latter is reserved for the subset of NICU infants in whom time to result is critical, either because of the severity of their illness or because of time constraints of medical decision making or significant interventions (such as starting extra-corporeal membrane oxygenation (ECMO)).
Figure 2.
Steps and minimum times of rapid genetic disease diagnosis by whole genome sequencing and implementation of precision neonatology. EHR, electronic health record.
Diagnostic genome sequencing requires two inputs. Firstly, the clinical features of the child’s illness are extracted from the medical record. This extraction is often performed computationally by natural language processing. The observed clinical features, typically as Human Phenotype Ontology (HPO) terms, are then compared computationally with the expected clinical features of all known genetic diseases to create a quantitatively prioritized, differential diagnosis list. Alternatively, the clinical features are used to select from a set of virtual gene panels. The second input is genome sequence from DNA extracted from 50 μL of peripheral blood. The DNA is then prepared for sequencing, a process called library preparation, which involves random fragmentation into ~500 nucleotide pieces, and attachment of short DNA probes on either end. Genome sequencing is then performed. For exome or gene panel sequencing, two additional steps, namely enrichment of exons and amplification of the remaining material, are performed. Sample preparation from blood sampling to start of sequencing takes from 2 hours to 2 days. The longer preparation time is required for exome and panel sequencing. Sequencing is almost always by synthesis, resulting in a pair of 100 – 150 nucleotide sequences (reads) from either end of each fragment. Genomes are sequenced to at least 30-fold coverage (~100 gigabases (GB) of DNA sequence), while exomes are sequenced to at least 90-fold coverage (~10 GB). Sequencing takes 11 hours – 2 days, depending on instrument settings, and is performed from single samples all the way to 50 samples per run. The remaining steps are computational and either performed in a cloud environment or on local high-performance computing instruments. First, the nucleotide of each position on each read is determined, together with a quality score. In general, quality must be better than 1 error in a 1,000 (Quality (Q) score of >30). Secondly, each pair of sequences is mapped to the corresponding, unique region of the genome (alignment), and a mapping quality score is determined. Thirdly, the aggregate sequence at each position is determined based on the consensus of ~30 reads in genome sequencing. This involves determining the DNA sequence, zygosity, and copy number. All nucleotides and regions that differ from the reference genome are identified (variant calling). In a genome, this is typically over 3.5 - 5 million single nucleotide substitutions (single nucleotide variants, SNVs), 750,000 – 1 million insertions and deletions of size 1 – 50 nucleotides (indels), and 20,000 structural and copy number variants that range in size from 50 nucleotides to entire chromosomes (Table 1). Variants are mapped to genes, genes to diseases, and diseases to the differential diagnosis calculated based on the infant’s clinical features. To diagnose a genetic disease, the search space is almost always limited to those genes. Pathogenicity prediction algorithms are used to predict the effect of each variant on gene or protein function. About 99% of variants are predicted not to affect function (Table 1). Mendelian Inheritance in Man (MIM) lists 7,036 genetic diseases that map to 4,545 genes (72). Next, due to the low frequencies of neonatal genetic diseases associated with purifying selection pressure, all variants that are common in populations are discarded. With a few exceptions, the cutoff is typically 1% minor allele frequency (MAF), removing 95% of variants (Table 1). These steps generate a short list of variants that are evaluated one by one, based on a large number of additional considerations, such as diplotype zygosity, allele frequency, computational pathogenicity prediction, and de novo occurrence. Each variant is compared with several reference databases of disease-causing and non-disease-causing variants. Dependent upon the patient’s age, location, such as the NICU, and clinical features, a genetic disease diagnosis is made in 10% to 50% of cases. In 5% of cases, two genetic diseases co-exist. In addition to findings that are considered diagnostic, in about 20% to 35%, variants of uncertain significance (VUSs) in genes associated with the phenotypic characteristics of the child’s presentation are identified. These are often called VUS-suspicious, and are typically also reported. In 5-10% of cases, there will be incidental findings (diagnostic findings that are considered unrelated to the child’s current illness). This process, genome interpretation, can be fully automated, taking 5-15 minutes, with retention of about 80% sensitivity and about 50% specificity, values which are currently insufficient to permit autonomous performance (20). Expert manual interpretation by specialist staff including physicians and molecular laboratory directors typically takes one hour to one day. Results likely to change management in a highly beneficial manner are typically returned immediately, verbally as a provisional report. About one third of results undergo orthogonal confirmatory testing to verify, for example, that two variants in a recessive condition are in trans, de novo status when parental genomic sequences are not available, or to exclude the possibility that variants are false positives.
Table 1.
Comparison of the median analytic performance of rWES (n=95) and rWGS (n=118). Wilcoxon signed-rank adjusted p-values
rWES or rWGS | % Coding Nt with ≥10X Coverage | Total Variants | SNVs | Indels | Coding variants | Rare Variants (MAF <1%) | Variants in MIM disease genes | Missense Variants | Non-sense Variants | Altered canonical splice sites | Frame-shift indels | Disrupted start codons |
---|---|---|---|---|---|---|---|---|---|---|---|---|
rWES | 94.5 | 38,901 | 35,465 | 3,401 | 23,421 | 2,703 | 670 | 558 | 14 | 15 | 46 | 3 |
rWGS | 98 | 4,669,310 | 3,792,213 | 881,699 | 26,080 | 240,648 | 48,231 | 687 | 16 | 82 | 85 | 4 |
Fold difference | 1.04 | 121 | 107 | 258 | 1.12 | 85 | 69 | 1.3 | 1.3 | 5.4 | 1.8 | 1.3 |
p-value | <.0001 | <.0001 | <.0001 | <.0001 | <.0001 | <.0001 | <.0001 | <.0001 | <.0001 | <.0001 | 0 |
Diagnostic and Clinical Utility of Rapid Genome Sequencing
The diagnostic yield of rapid genome sequencing in infants and children in ICUs was evaluated in 33 clinical studies between 2012 and November 2021 (Table 2) (3, 13, 16, 21, 24, 25, 27, 28, 32, 43, 63, 64, 73, 79, 82–84, 86, 87, 96–98, 103, 109, 110). Most were cohort studies. Fifteen evaluated rWGS, 18 rWES, and one rapid panel testing. While the inclusion criteria, clinical settings, and hospital systems varied, all evaluated the diagnostic yield in children in ICUs with suspected genetic diseases. Average turnaround time varied from 0.8 days to 60 days. The weighted average rate of genetic disease diagnosis was 36% (range 19% - 83%, n=2874). Twenty-nine studies (n=1037) also evaluated acute clinical utility, as measured by changes in management upon return of results. The weighted average rate of change in management was 27% (range 7% - 60%). Ten studies (n=487) examined changes in outcome following those changes in management. The weighted average rate of change in outcome was 19% (range 8% - 30%).
Table 2.
Studies of the diagnostic performance, clinical utility and change in outcome of rWES, rWGS and rapid panel tests in seriously ill children in intensive care units
Ref. | Year | Study Type | Test Type | Enrollment Criteria | Size | Dx Rate | Change in Management | Change in Outcome | TAT (d) |
---|---|---|---|---|---|---|---|---|---|
83 | 2012 | Cases | urWGS | NICU infants with suspected genetic disease | 4 | 75% | n.d. | n.d. | 2 |
103 | 2015 | Cohort | rWGS | <4 mo of age; Suspected actionable genetic disease | 35 | 57% | 31% | 29% | 23 |
63 | 2017 | Cohort | rWES | <100 days of life; Suspected genetic disease | 63 | 51% | 37% | 19% | 13 |
96 | 2017 | Cohort | rWGS | Infants; NICU, PICU; susp. genetic diseases | 23 | 30% | 22% | 22% | 12 |
64 | 2018 | RCT | rWGS, SOC | <4 mo of age; Suspected genetic disease | 32 | 41% | 31% | n.d. | 13 |
25 | 2018 | Cohort | rWGS | infants; Suspected genetic disease | 42 | 43% | 31% | 26% | 23 |
92 | 2018 | Cohort | rWES | Acutely ill children with suspected genetic diseases | 40 | 53% | 30% | 8% | 16 |
90 | 2018 | Cohort | rWGS | Children; PICU and Cardiovascular ICU | 24 | 42% | 13% | n.d. | 9 |
82 | 2019 | Cohort | rWGS | 4 months-18 years; PICU; Suspected genetic diseases | 38 | 48% | 39% | 8% | 14 |
28 | 2019 | Cohort | rWGS | Suspected genetic disease | 195 | 21% | 13% | n.d. | 21 |
17 | 2019 | Cases | urWGS | Infants; ICU; Suspected genetic disease | 7 | 43% | 43% | n.d. | 0.8 |
32 | 2020 | Cohort | rWES | <6 mo; ICU; hypotonia, seizures, metabolic, multiple congenital anomalies | 50 | 58% | 48% | n.d. | 5 |
24 | 2019 | Cohort | rWES | NICU; infants; susp. genetic disease | 25 | 72% | 60% | n.d. | 7.2 |
110 | 2019 | Cohort | rWES | PICU and other; children; susp. genetic disease | 40 | 53% | 43% | n.d. | 6 |
97 | 2020 | Cohort | rWES | NICU & PICU; complex | 130 | 48% | 23% | n.d. | 3.8 |
27 | 2020 | Cohort | rWES | Critical illness; medical genetics selected | 46 | 43% | 52% | n.d. | 9 |
13 | 2020 | Cohort | rWES | PICU; < 6 years; new metabolic/neurologic disease | 10 | 50% | 30% | n.d. | 9.8 |
87 | 2020 | Cohort | rWES | ICU | 368 | 27% | n.d. | n.d. | n.d. |
16 | 2020 | Cohort | rWES | >1 year; ICU and inpatient | 102 | 31% | 27% | n.d. | 11 |
79 | 2020 | Cohort | rWES | Various | 41 | 32% | n.d. | n.d. | 7 |
3 | 2020 | Implem | rWES | <18 yr; NICU and PICU | 108 | 51% | 44% | n.d. | 3 |
86 | 2020 | Cohort | rWES | Infants; NICU, PICU; susp. genetic diseases | 18 | 83% | 61% | n.d. | 14 |
98 | 2020 | Cohort | rWES | Infants; NICU, PICU; susp. genetic diseases | 33 | 70% | 30% | 30% | 1 |
rWGS | 94 | 19% | 24% | 10% | 11 | ||||
11, 23, 43 | 2019-2021 | RCT | rWES | Infants; disease of unknown etiology; with in 96 hours of admission | 95 | 20% | 20% | 18% | 11 |
urWGS | 24 | 46% | 63% | 25% | 4.6 | ||||
58 | 2021 | Crossover | rWGS, panel | Infants; disease of unknown etiology | 113 | 33% | 26% | n.d. | n.d. |
22 | 2021 | Implem | rWGS | Medicaid infants; unknown etiology; within 1 week of admission | 178 | 43% | 31% | n.d. | 3 |
73 | 2021 | Cohort | rWES | Critically ill; 6 days-15 years; susp. genetic diseases | 40 | 43% | 31% | n.d. | 5 |
84 | 2021 | Cohort | rWES | NICU, PICU, infants; sup. Genetic diseases | 61 | 43% | 11% | n.d. | 60 |
77 | 2021 | RTDCT | rWGS, WGS | 0-120 days old; ICU; susp. genetic disease | 354 | 31% | 25% | n.d. | 15 |
109 | 2021 | Crossover | rWES | Critically ill infants with conditions suggestive of geneticaly heterogeneous disorders | 202 | 20% | n.d. | n.d. | 20 |
rWGS | 202 | 37% | 7% | n.d. | 7 | ||||
21 | 2021 | Cohort | rWGS | NICU, PICU units with probable genetic disease and in urgent need for etiological diagnosis to guide medical care | 37 | 57% | n.d. | n.d. | 43 |
| |||||||||
Weighted Average | 2874 | 36% | 27% | 18% |
n.d.: not done; d: days
A meta-analysis compared the diagnostic and clinical utility of WGS, WES, and CMA in 20,068 children with suspected genetic diseases through August 2017 (17). It found that the diagnostic utility of WGS (41%, 95% CI 34-48%) and WES (36%, 95% CI 33-40%) was not significantly different, but was greater than CMA (10%, 95% CI 8-12%). Diagnosis was significantly more likely for trios than singletons (odds ratio 2.04, 95% CI 1.62-2.56). Interestingly, hospital-based interpretation led to a higher rate of diagnosis by WGS/WES (42%, 95% CI 38-45%) than that of reference laboratories (29%, 95% CI 27-31%). WGS results had greater clinical utility (27%, 95% CI 17-40%) than CMA (6%, 95% CI 5-7%).
There have been three randomized, controlled trials (RCTs) of rapid genome sequencing in infants in ICUs. The first, Newborn Sequencing in Genomic Medicine and Public Health (NSIGHT1), compared the rate of genetic disease diagnosis with rWGS plus standard genetic tests to that of standard genetic tests alone (including exome sequencing) in 65 infants aged <4 months in a regional NICU and pediatric intensive care unit (PICU) with illnesses of unknown etiology (77). The study was terminated early due to loss of equipoise: 15% of controls underwent compassionate cross-over to receive rWGS, demonstrating the difficulty of diagnostic RCTs. The rate of genetic diagnosis within 28 days of enrollment (the primary end point) was higher with rWGS (31%) than controls (3%). Median time to diagnosis was significantly less with rWGS (13 days) than controls (107 days). This study established that rWGS increased the proportion of NICU/PICU infants who received timely diagnoses of genetic diseases relative to standard genetic tests.
The second RCT, NSIGHT2, evaluated effectiveness of rWES, rWGS, and urWGS as first-tier tests in 213 seriously ill infants with diseases of unknown etiology, representing 46% of NICU admissions (a broader proportion than previously studied) (43). The analytic performance of rWGS/urWGS was superior to rWES (Table 1). Their diagnostic rates and times to result of rWGS and rWES were not different (~20% and median of 11 days, respectively). Both, however, were inferior to urWGS (46% diagnosis, median time to result 4.6 days). The incremental diagnostic yield of reflexing to trio after negative singleton analysis was only 0.7%, a result that was at odds with the previously published meta-analysis. A second report from the NSIGHT2 RCT demonstrated that clinicians perceived rapid genome sequencing to be useful (the primary NSIGHT2 study end point) in 77% of infants (Figure 3) (23). Interestingly, both positive (93%) and negative tests (72%) had clinical utility. The explanation of the latter was that negative rapid genome sequencing results were useful in decreasing the posterior probability of genetic disease, enabling clinicians to focus on non-genetic etiologies. Results of rapid genome sequencing changed clinical management in 28% infants and outcomes in 15%.
Figure 3.
Comparison of the traditional approach to etiologic diagnosis of genetic diseases in NICU infants with rWGS-informed precision medicine. Values are from the NSIGHT2 RCT (11, 23, 43)
A third report from the NSIGHT2 RCT evaluated parental perceptions of clinical utility, adequacy of consent, and potential harms and benefits (Figure 3) (11). When rWGS was first introduced, there were concerns that parents of newborns in ICUs would be unable to provide informed consent and that testing would cause anxiety, be associated with decisional regret, depression, and lead to interference with bonding. However, more than 90% of NICU infant parents felt adequately informed to consent to diagnostic genome sequencing. Despite only 23% of infants’ receiving diagnoses, 97% of parents reported that genome sequencing was useful, and median decisional regret was zero (range 0-100). Evaluation of potential harms of testing revealed that only 2% perceived harm (1% related to a negative result and 2% related to stress or confusion). This study established that when rapid genome sequencing was performed as a first-tier diagnostic test in almost one half of regional NICU infants, most results were considered useful, and harms were rare and mild.
The third RCT was a multicenter, time-delayed trial of 354 infants in ICUs with suspected genetic disease aged <4 months who were randomized to receive WGS results within either 15 or 60 days after enrollment (31). Among infants who received WGS results within 15 days, 31% had diagnoses, and 21% had consequent changes in management at 60 days after enrollment, compared with 15% and 10%, respectively, among those who received results within 60 days. This study established the superiority of use of rWGS as a first-tier diagnostic test.
In summary, research studies have established an evidence base for the diagnostic and clinical utility of rWGS as a first tier diagnostic test for NICU infants with diseases of unknown etiology. This evidence base contributed to publication by the ACMG of a clinical guideline that supports the clinical utility and desirable effects of WES and WGS on active and long-term clinical management for pediatric patients <1 year of age with 1 or more congenital anomalies (56).
Implementation Science and Quality Improvement
While research studies are ideal for testing hypotheses, they feature biases that can lead to their results’ failing to be repeated in general experience. Inclusion and exclusion criteria and requirement for informed parental consent, for example, can result in enrollment that is not representative of NICU populations. Furthermore, research studies employ staff such as genome-literate research nurses and genetic counselors who may upskill NICU teams, enabling practices that would not be possible in their absence. Translation of novel, evidence-based practices into routine clinical use typically takes decades. The field of implementation science has developed strategies to facilitate adoption of evidence-based practices (60, 78, 101, 102). It is defined as “the scientific study of methods to promote the systematic uptake of research findings and other evidence-based practices into routine practice, and, hence, to improve the quality and effectiveness of health services” (5). Implementation science involves comprehensive identification of the key barriers to adoption, their organization within a framework, and then real-world implementation studies that explore how to refine implementation to optimize effectiveness and facilitate adoption. Sites in the US, UK, and Australia started implementation science studies of rapid genome sequencing in 2017 motivated by the desire to scale use by NICU teams and achieve sustainable improvement in outcomes (14). The resultant framework developed by Rady Children’s Institute for Genomic Medicine (RCIGM) is shown in Figure 4 (45). Assessment of barriers to adoption across ~70 NICUs served by RCIGM revealed the need to implement rWGS within a learning, rapid precision medicine delivery system (RPM). Based on the successful framework for implementation of traditional newborn screening (8), RPM had four components: 1. Onboarding of health systems, upskilling of NICU teams, and early identification of infants in need during admission (items 1-7, Figure 4); 2. Diagnosis, comprising rWGS ordering, sequencing, interpretation and results review with ordering pediatricians (items 8-13, Figure 5); 3. Acute management guidance and precision medicine delivery (items 15 and 16, Figure 5), and 4. Learning based on monitoring outcomes, data science and quality improvement (items 14, 17 and 18, Figure 4) (45). Implementation science studies of rapid genome sequencing have started to be published. For example, Project Baby Bear, a payer-funded, implementation project, evaluated the clinical and economic impact of the RCIGM system of rWGS-based RPM (22, 26). Rapid WGS was utilized as a first line diagnostic test in Medicaid infants with diseases of unknown etiology in five regional California NICUs. The majority of infants were from underserved populations. Of 184 infants enrolled, 40% received a diagnosis by rWGS that explained their admission with a median turnaround time of 3 days. In 32% of infants, rWGS led to changes in medical care. rWGS testing and resultant precision medicine cost $1.7 million (~$21,000 per diagnosis) but led to ~$2.5 million cost savings ($13,526 per infant tested, Figure 5). Sensitivity analysis showed that most of the cost savings were lost if turnaround time was lengthened to 14 days. Thus, Project Baby Bear confirmed the diagnostic and clinical utility previously shown in research studies and demonstrated net reduction in healthcare utilization costs associated with relatively broad indications for first tier rWGS. Analysis of the implementation process revealed the need for an rWGS champion in each NICU, educational needs and strategies, negotiating decision-making roles and processes, workflows and workarounds, and perceptions about rWGS. A similar implementation pilot in 12 hospitals demonstrated the feasibility of rWES with 3-day turnaround time in critically ill pediatric patients with suspected monogenic conditions in the Australian public health care system (3). Genetic diseases were diagnosed in 51% of infants and management was changed in 76% of those receiving diagnoses and 11% of infants with negative reports. An earlier implementation project by the same group in two Australian centers showed similar rates of diagnostic and clinical utility, while the cost per diagnosis was $10,453, and net cost savings were $10,600 per infant tested (90).
Figure 4.
Genome-informed neonatology is a learning healthcare delivery system with four components: 1. Education, engagement, and equipping (yellow line); 2. Rapid, diagnostic whole genome sequencing (green line); 3. Translation into precision neonatology (blue line); 4. Therapeutic innovation (blue line). Learning feedback loops are shown.
Figure 5:
Cost effectiveness of precision neonatology is dependent on time to result. a. Cost savings during the initial hospitalization from first-tier use of rWGS for NICU infants with suspected genetic diseases in Project Baby Bear. b. Total cost savings.
Policies and Guidelines
Three national health systems (the National Health Service in England and in Wales and Australian Genomics) are adopting rWGS in critically ill, inpatient infants with suspected genetic diseases. In the US, California Blue Shield was the first major payer to issue a coverage policy for rapid, diagnostic WES/WGS in NICU infants and trios in July 2019 (Box 1) (12). Rapid testing was defined as an average turnaround time of less than 14 days, but usually less than 7 days. The policy called for immediate verbal reporting of results to the clinician if changes in management were likely. The criteria for reimbursable testing are shown in Box 1 and reflect those used in published research studies such as Project Baby Bear and NSIGHT2. A new current procedural terminology (CPT) code was authorized for rWGS (0094U). In March 2020, the policy was accepted by Anthem/Blue Cross/Blue Shield of America and has since been ratified by ten regional plans. In September 2021, Michigan became the first US state to reimburse rWGS in critically ill Medicaid infants in NICUs and PICUs as a carve-out from the intensive care diagnosis related codes (DRGs). The Michigan coverage policy is very similar to that of Blue Cross/Blue Shield. The CPT code 0094U was implemented and priced at $6,275 for probands and $10,750 for trios. In July 2021, Governor
Box 1. Policy for coverage of rWGS and rWES in infants in ICUs by Blue Shield California.
Rapid whole exome sequencing or rapid whole genome sequencing, with trio testing when possible, meets the definition of medical necessity for the evaluation of critically ill infants in neonatal or pediatric intensive care with a suspected genetic disorder of unknown etiology when both (1 & 2) of the following criteria are met:
- At least one of the following criteria is met:
- Multiple congenital anomalies (e.g. persistent seizures, abnormal ECG, hypotonia);
- An abnormal laboratory test or clinical features suggests a genetic disease or complex metabolic phenotype (e.g, abnormal newborn screen, hyperammonemia, lactic acidosis not due to poor perfusion); or
- An abnormal response to standard therapy for a major underlying condition.
- None of the following criteria apply regarding the reason for admission to intensive care:
- An infection with normal response to therapy;
- Isolated prematurity;
- Isolated unconjugated hyperbilirubinemia;
- Hypoxic Ischemic Encephalopathy:
- Confirmed genetic diagnosis explains illness;
- Isolated Transient Neonatal Tachypnea;
- Nonviable neonates.
Newsom signed California Assembly Bill 114 into law, providing $6 million to reimburse rWGS in Medicaid infants in California in 2022. A coverage policy has not yet been issued by the California Department of Health. Feasibility projects of rapid WGS and WES are underway in NICU infants in many high and middle income countries (89).
Future Directions
Many research and implementation studies are currently underway that will continue to refine our understanding of the use of genome sequencing in NICUs and in other pediatric intensive care settings (Pediatric Intensive Care Units and Pediatric Cardiac Intensive Care Units). It is already apparent, however, that rWGS-based precision medicine has considerable diagnostic and clinical utility and cost effectiveness in infants in ICUs. This evidence will drive an era of broad adoption of rWGS-informed precision medicine for infants in NICUs. We anticipate issuance of guidelines from professional bodies that support use of rWGS as a first-tier diagnostic test in critically ill infants with diseases of unknown etiology. We anticipate that guidelines will recommend testing shortly after admission, turnaround time of 3 days, and reports to include all classes of variants. We anticipate rWGS-informed precision medicine will become part of the core curriculum for neonatology fellowship. In parallel, there will continue to be broader reimbursement of rWGS by Medicaid and private payers. It will be important for coverage policies to include reimbursement as a carve-out payment rather than inclusion in current DRGs.
A key unanswered question at present is how broadly genome sequencing should be used in NICUs. The NSIGHT2 study showed diagnostic and clinical utility when genomic sequencing was used in 46% of admissions to a regional NICU. It was the first to quantify the value of negative genomic sequencing. Further studies are needed to examine optimal breadth of use in level II, III, and IV NICUs.
The research published to date still underestimates the prevalence of single locus genetic diseases in infants in ICUs. An unpublished study of postmortem infants from one of us found a high rate of undiagnosed genetic disorders, many of which had effective treatments in a county with relatively broad use of NICU and PICU rWGS (45, 81). Genetic diseases continue to be discovered at a rapid pace, and the quality of WGS continues to improve rapidly. For example, it is likely that diagnostic yield will increase by 5 – 15% through use of combined long-and short-read WGS (65). Long-read WGS is more expensive than short-read WGS. It is not as good at detection of SNVs but far superior for characterization of SVs and CNVs. The emerging picture of these variants is that they are generally more complex than we were aware and frequently are combination events that include deletions, insertions, and rearrangements at a single locus. Also in the relatively near future, we will change from read alignment to read assembly, generating assembled individual genomes (85). It is likely that this will increase yield by another 5 – 15%, particularly in racial and ethnic groups that did not contribute to the current reference human genome. An intermediary to genome assemblies may be digital, automated ethnic ancestry delineation in short-read WGS and then alignment to the best from a large set of diverse reference genomes (38). Lastly, as we enter a new era of integrative, clinical omics, we are starting to see the value of integrating WGS with functional omics, such as RNAseq (51), proteomics, and metabolomics. Currently, there are a very large number of variants for which we cannot predict pathogenicity. Most of these are intergenic or intronic. These omics technologies allow us to evaluate the functional consequences of such variants. The two uses of such technology are to diagnose unsolved cases and to build new databases of functional omics-annotated variants. Ultimately, there will be very few remaining VUSs.
Another trend that is gaining momentum is the use of artificial intelligence (AI) to improve scalability and increase adoption of rapid genome sequencing and precision medicine in infants in ICUs. In addition to the emerging uses of AI discussed previously, we anticipate algorithm-based, automated EHR alerts triggered in infants with high likelihood of underlying genetic disease and at high risk of mortality (akin to EHR sepsis alerts) (80). An unpublished study from one of us (SFK) describes the development of an AI-informed, automated system for provision of acute management guidance for newly diagnosed genetic diseases. It was designed for use by front-line intensivists and neonatologists, particularly in hospitals that lack a full array of subspecialists and super-specialists where there may be delays in implementation of optimal treatments for ultra-rare genetic diseases. AI tools such as these will be critical in democratization of rWGS-based precision medicine to most birthing-hospital associated NICUs.
The most exciting future development will be accelerated development of genetic therapies (Figure 1). Effective gene therapy for Spinal Muscular Atrophy type 1 (SMA1) is a staggering accomplishment. We are realizing, however, that the system shown in Figure 4 is critically needed in order to diagnose and treat hypotonic babies in the first week of life. A byproduct of increasing use of rWGS is unparalleled natural history studies of specific infant-onset genetic diseases that accelerate drug development both by identification of end-points and indications and by serving as real-world evidence for submissions investigational new drug applications to the Food and Drug Administration (FDA) in the US (1). It must be understood that the majority of infant-onset genetic diseases was either only recently discovered or was not generally diagnosable in the absence of rapid genome sequencing. Thus, many genetic diseases for which drug development was not previously feasible now have potential therapeutic strategies, including gene therapy, anti-sense oligonucleotides, small molecule, genome editing, and repurposed FDA approved drugs. In addition to better therapies, natural history studies will improve our ability to predict prognosis for critically ill infants, communicate with parents, anticipate complications, and stratify patients within genetic disorders as is now happening with Duchenne Muscular Dystrophy and Cystic Fibrosis.
Finally, and beyond the scope of this review, is newborn screening for genetic diseases by rWGS (NBS-by-WGS) (9) and increasing use of rWGS for fetal diagnosis. We anticipate that healthy newborns will start to be widely screened by WGS for ~500 infant onset, severe genetic diseases that have effective treatments within the next five years. Professional society opinions support the use of prenatal exome sequencing in cases with specific fetal anomalies, and we anticipate imminent testing of fetal rWGS to improve diagnostic success (18, 35, 69, 94, 99)
Next steps: broad implementation of genome sequencing and associated research –
Genetic heterogeneity of fetal and neonatal phenotypes, overrepresentation of novel and rare genomic variants in the NICU population, the high fraction of affected infants in NICUs with undiagnosed congenital anomalies or metabolic disorders, the significant contribution of genetic disorders to morbidity and mortality among infants, and the proven record of rWGS/rWES to change clinical management and identify individualized therapeutic strategies make the integration of genome sequencing into NICU clinical care a high priority (101, 102). As illustrated by the framework developed by Rady Children’s Institute for Genomic Medicine (RCIGM) described above (45), acceleration of this integration will require an intentional commitment to implementation of a sustainable Genomic Learning Healthcare System (GLHS) through implementation science which uses knowledge and experience derived from NICU and prenatal clinical care for cycles of continuous improvement and research to improve patient outcomes through enhanced diagnostic success, clinical efficiency, and discovery of genotype-guided therapeutics (60–62, 101, 102, 108). Multiple stakeholders including parents, payers, bioinformaticians, geneticists, genetic counselors, neonatologists, obstetricians, pediatric subspecialists, developmental biologists, model organism investigators, experts in strategies to rescue variant-encoded disruption (e.g., with gene therapy, anti-sense oligonucleotides, repurposed drugs approved by the FDA, small molecules, or genome editing), clinical investigators, genomicists, and institutional leaders will need to participate in development of the GLHS for NICUs. The challenges to GLHS implementation include facilitating a cultural shift among providers from phenotype- to genotype-first diagnosis, development of governance structures that can adapt to new ethical and operational questions, standardization of genomic and phenotypic information in EHRs, more reliable computational and functional evaluation of novel and rare VUSs that are clinically actionable, and development of consent strategies that permit inclusion of an individual’s clinical and genomic data while protecting patient confidentiality (61, 100, 108). However, NICUs have a long record of commitment to quality improvement through implementation science and, more recently, of value-based quality initiatives (33). With the already available evidence of clinical value of r- and ur-WES and WGS in the NICU, GLHS implementation will lead to deployment of these best practices to improve outcomes for critically ill newborn infants and children and reduce costs of their care.
Terms and Definitions:
- ACMG
American College of Medical Genetics and Genomics
- AI
Artificial intelligence
- BNS1
Benign neonatal seizures type 1
- CDH
Congenital diaphragmatic hernia
- CMA
Chromosomal microarray analysis
- CPT
Current procedural terminology
- DEE7
Developmental and epileptic encephalopathy type 7
- DRG
Diagnosis related code
- ECMO
Extracorporeal membrane oxygenation
- FDA
Food and Drug Administration
- GB
Gigabase
- GINA
Genetic Information Nondiscrimination Act
- GLHS
Genomic Learning Healthcare System
- HPO
Human Phenotype Ontology
- Indel
Insertion and deletion
- MAF
Minor allele frequency
- MIM
Mendelian Inheritance in Man
- NICU
Neonatal Intensive Care Unit
- NSIGHT
Newborn Sequencing in Genomic Medicine and Public Health
- PICU
Pediatric intensive care unit
- RCT
Randomized controlled trial
- RPM
Rapid precision medicine
- rWES
Rapid whole exome sequencing
- rWGS
Rapid whole genome sequencing
- SNV
Single nucleotide variant
- TAT
Turnaround time
- THMD2
Thiamine metabolism dysfunction syndrome 2
- urWGS
Ultra-rapid whole genome sequencing
- VUS
Variant of uncertain significance
- WES
Whole exome sequencing
- WGS
Whole genome sequencing
Contributor Information
Stephen F. Kingsmore, Rady Children’s Hospital Institute for Genomic Medicine, Rady Children’s Hospital-San Diego
F. Sessions Cole, Division of Newborn Medicine, Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine in St. Louis
Literature Cited
- 1.Administration USFD. 2021. Real-world data (RWD) and real-world evidence (RWE) are playing an increasing role in health care decisions. In Real-World Evidence, ed. FDA. FDA website: FDA [Google Scholar]
- 2.Almli LM, Ely DM, Ailes EC, Abouk R, Grosse SD, et al. 2020. Infant Mortality Attributable to Birth Defects - United States, 2003-2017. MMWR Morb Mortal Wkly Rep 69: 25–29 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Australian Genomics Health Alliance Acute Care F, Lunke S, Eggers S, Wilson M, Patel C, et al. 2020. Feasibility of Ultra-Rapid Exome Sequencing in Critically Ill Infants and Children With Suspected Monogenic Conditions in the Australian Public Health Care System. JAMA 323: 2503–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Bainbridge MN, Wiszniewski W, Murdock DR, Friedman J, Gonzaga-Jauregui C, et al. 2011. Whole-genome sequencing for optimized patient management. Sci Transl Med 3: 87re3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bauer MS, Damschroder L, Hagedorn H, Smith J, Kilbourne AM. 2015. An introduction to implementation science for the non-specialist. BMC Psychol 3: 32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bayat A, Bayat M, Rubboli G, Moller RS. 2021. Epilepsy Syndromes in the First Year of Life and Usefulness of Genetic Testing for Precision Therapy. Genes (Basel) 12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Bell CJ, Dinwiddie DL, Miller NA, Hateley SL, Ganusova EE, et al. 2011. Carrier testing for severe childhood recessive diseases by next-generation sequencing. Sci Transl Med 3: 65ra4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Berry SA. 2015. Newborn screening. Clin Perinatol 42: 441–53, x [DOI] [PubMed] [Google Scholar]
- 9.Biesecker LG, Green ED, Manolio T, Solomon BD, Curtis D. 2021. Should all babies have their genome sequenced at birth? BMJ 375: n2679. [DOI] [PubMed] [Google Scholar]
- 10.Brynn L 2018. Prenatal diagnosis. In Methods in Molecular Biology, ed. Walker JM. New York, New York: Humana Press [Google Scholar]
- 11.Cakici JA, Dimmock DP, Caylor SA, Gaughran M, Clarke C, et al. 2020. A Prospective Study of Parental Perceptions of Rapid Whole-Genome and -Exome Sequencing among Seriously Ill Infants. Am J Hum Genet 107: 953–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Calirofnia BSo. 2021. Whole Exome and Whole Genome Sequencing for Diagnosis of Genetic Disorders. In Medical Policy, ed. BSo California, pp. Medical policy. Blue Shield website: Blue Shield of California [Google Scholar]
- 13.Carey AS, Schacht JP, Umandap C, Fasel D, Weng C, et al. 2020. Rapid exome sequencing in PICU patients with new-onset metabolic or neurological disorders. Pediatr Res 88: 761–68 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Chambers DA, Feero WG, Khoury MJ. 2016. Convergence of Implementation Science, Precision Medicine, and the Learning Health Care System: A New Model for Biomedical Research. JAMA 315: 1941–2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Choi M, Scholl UI, Ji W, Liu T, Tikhonova IR, et al. 2009. Genetic diagnosis by whole exome capture and massively parallel DNA sequencing. Proc Natl Acad Sci U S A 106: 19096–101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Chung CCY, Leung GKC, Mak CCY, Fung JLF, Lee M, et al. 2020. Rapid whole-exome sequencing facilitates precision medicine in paediatric rare disease patients and reduces healthcare costs. Lancet Reg Health West Pac 1: 100001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Clark MM, Stark Z, Farnaes L, Tan TY, White SM, et al. 2018. Meta-analysis of the diagnostic and clinical utility of genome and exome sequencing and chromosomal microarray in children with suspected genetic diseases. NPJ Genom Med 3: 16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Committee on G, the Society for Maternal-Fetal M. 2016. Committee Opinion No.682: Microarrays and Next-Generation Sequencing Technology: The Use of Advanced Genetic Diagnostic Tools in Obstetrics and Gynecology. Obstet Gynecol 128: e262–e68 [DOI] [PubMed] [Google Scholar]
- 19.Conley ME, Dobbs AK, Farmer DM, Kilic S, Paris K, et al. 2009. Primary B cell immunodeficiencies: comparisons and contrasts. Annu Rev Immunol 27: 199–227 [DOI] [PubMed] [Google Scholar]
- 20.De La Vega FM, Chowdhury S, Moore B, Frise E, McCarthy J, et al. 2021. Artificial intelligence enables comprehensive genome interpretation and nomination of candidate diagnoses for rare genetic diseases. Genome Med 13: 153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Denomme-Pichon AS, Vitobello A, Olaso R, Ziegler A, Jeanne M, et al. 2021. Accelerated genome sequencing with controlled costs for infants in intensive care units: a feasibility study in a French hospital network. Eur J Hum Genet [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Dimmock D, Caylor S, Waldman B, Benson W, Ashburner C, et al. 2021. Project Baby Bear: Rapid precision care incorporating rWGS in 5 California children’s hospitals demonstrates improved clinical outcomes and reduced costs of care. Am J Hum Genet [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Dimmock DP, Clark MM, Gaughran M, Cakici JA, Caylor SA, et al. 2020. An RCT of Rapid Genomic Sequencing among Seriously Ill Infants Results in High Clinical Utility, Changes in Management, and Low Perceived Harm. Am J Hum Genet 107: 942–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Elliott AM, du Souich C, Lehman A, Guella I, Evans DM, et al. 2019. RAPIDOMICS: rapid genome-wide sequencing in a neonatal intensive care unit-successes and challenges. Eur J Pediatr 178: 1207–18 [DOI] [PubMed] [Google Scholar]
- 25.Farnaes L, Hildreth A, Sweeney NM, Clark MM, Chowdhury S, et al. 2018. Rapid whole-genome sequencing decreases infant morbidity and cost of hospitalization. NPJ Genom Med 3: 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Franck LS, Kriz RM, Rego S, Garman K, Hobbs C, Dimmock D. 2021. Implementing Rapid Whole-Genome Sequencing in Critical Care: A Qualitative Study of Facilitators and Barriers to New Technology Adoption. J Pediatr 237: 237–43 e2 [DOI] [PubMed] [Google Scholar]
- 27.Freed AS, Clowes Candadai SV, Sikes MC, Thies J, Byers HM, et al. 2020. The Impact of Rapid Exome Sequencing on Medical Management of Critically Ill Children. J Pediatr [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.French CE, Delon I, Dolling H, Sanchis-Juan A, Shamardina O, et al. 2019. Whole genome sequencing reveals that genetic conditions are frequent in intensively ill children. Intensive Care Med 45: 627–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Fridriksdottir R, Jonsson AJ, Jensson BO, Sverrisson KO, Arnadottir GA, et al. 2021. Sequence variants in malignant hyperthermia genes in Iceland: classification and actionable findings in a population database. Eur J Hum Genet 29: 1819–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Goodman DC LG, Harrison WN, Moen A, Mowitz ME, Ganduglia Cazaban C, Bronner, (Eds.). KaDJ. 2019. The Dartmouth Atlas of Neonatal Intensive Care. Lebanon, New Hampshire: The Dartmouth Institute of Health Policy & Clinical Practice, Geisel School of Medicine at Dartmouth; [PubMed] [Google Scholar]
- 31.Group NIS, Krantz ID, Medne L, Weatherly JM, Wild KT, et al. 2021. Effect of Whole-Genome Sequencing on the Clinical Management of Acutely Ill Infants With Suspected Genetic Disease: A Randomized Clinical Trial. JAMA Pediatr [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Gubbels CS, VanNoy GE, Madden JA, Copenheaver D, Yang S, et al. 2020. Prospective, phenotype-driven selection of critically ill neonates for rapid exome sequencing is associated with high diagnostic yield. Genet Med 22: 736–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hagadorn JI, Johnson KR, Hill D, Sink DW. 2020. Improving the quality of quality metrics in neonatology. Semin Perinatol 44: 151244. [DOI] [PubMed] [Google Scholar]
- 34.Harrison W, Goodman D. 2015. Epidemiologic Trends in Neonatal Intensive Care, 2007-2012. JAMA Pediatr 169: 855–62 [DOI] [PubMed] [Google Scholar]
- 35.Hays T, Wapner RJ. 2021. Genetic testing for unexplained perinatal disorders. Curr Opin Pediatr 33: 195–202 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Heron M 2021. Deaths: Leading Causes for 2019. Natl Vital Stat Rep 70: 1–114 [PubMed] [Google Scholar]
- 37.Institute NHGR. 2021. DNA Sequencing Costs: Data. In About Genomics, ed. NHGRI, pp. Webpage. NHGRI website: NHGRI [Google Scholar]
- 38.Ioannidis AG, Blanco-Portillo J, Sandoval K, Hagelberg E, Barberena-Jonas C, et al. 2021. Paths and timings of the peopling of Polynesia inferred from genomic networks. Nature 597: 522–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Jensen K, Murray F. 2005. Intellectual property. Enhanced: intellectual property landscape of the human genome. Science 310: 239–40 [DOI] [PubMed] [Google Scholar]
- 40.Katsanis N 2016. The continuum of causality in human genetic disorders. Genome Biol 17: 233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kilby MD. 2021. The role of next-generation sequencing in the investigation of ultrasound-identified fetal structural anomalies. BJOG 128: 420–29 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kim JI, Ju YS, Park H, Kim S, Lee S, et al. 2009. A highly annotated whole-genome sequence of a Korean individual. Nature 460: 1011–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Kingsmore SF, Cakici JA, Clark MM, Gaughran M, Feddock M, et al. 2019. A Randomized, Controlled Trial of the Analytic and Diagnostic Performance of Singleton and Trio, Rapid Genome and Exome Sequencing in Ill Infants. Am J Hum Genet 105: 719–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kingsmore SF, Henderson A, Owen MJ, Clark MM, Hansen C, et al. 2020. Measurement of genetic diseases as a cause of mortality in infants receiving whole genome sequencing. NPJ Genom Med 5: 49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kingsmore SF, Ramchandar N, James K, Niemi AK, Feigenbaum A, et al. 2020. Mortality in a neonate with molybdenum cofactor deficiency illustrates the need for a comprehensive rapid precision medicine system. Cold Spring Harb Mol Case Stud 6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kingsmore SF, Saunders CJ. 2011. Deep sequencing of patient genomes for disease diagnosis: when will it become routine? Sci Transl Med 3: 87ps23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kitagawa H, Pringle KC. 2017. Fetal surgery: a critical review. Pediatr Surg Int 33: 421–33 [DOI] [PubMed] [Google Scholar]
- 48.Kong A, Frigge ML, Masson G, Besenbacher S, Sulem P, et al. 2012. Rate of de novo mutations and the importance of father’s age to disease risk. Nature 488: 471–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kosova G, Abney M, Ober C. 2010. Colloquium papers: Heritability of reproductive fitness traits in a human population. Proc Natl Acad Sci U S A 107 Suppl 1: 1772–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Krstic N, Obican SG. 2020. Current landscape of prenatal genetic screening and testing. Birth Defects Res 112: 321–31 [DOI] [PubMed] [Google Scholar]
- 51.Lee H, Huang AY, Wang LK, Yoon AJ, Renteria G, et al. 2020. Diagnostic utility of transcriptome sequencing for rare Mendelian diseases. Genet Med 22: 490–99 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Lee JS, Yoo T, Lee M, Lee Y, Jeon E, et al. 2020. Genetic heterogeneity in Leigh syndrome: Highlighting treatable and novel genetic causes. Clin Genet 97: 586–94 [DOI] [PubMed] [Google Scholar]
- 53.Levy S, Sutton G, Ng PC, Feuk L, Halpern AL, et al. 2007. The diploid genome sequence of an individual human. PLoS Biol 5: e254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lialiaris T, Mantadakis E, Kareli D, Mpountoukas P, Tsalkidis A, Chatzimichail A. 2010. Frequency of genetic diseases and health coverage of children requiring admission in a general pediatric clinic of northern Greece. Ital J Pediatr 36: 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Malfait F, Castori M, Francomano CA, Giunta C, Kosho T, Byers PH. 2020. The Ehlers-Danlos syndromes. Nat Rev Dis Primers 6: 64. [DOI] [PubMed] [Google Scholar]
- 56.Manickam K, McClain MR, Demmer LA, Biswas S, Kearney HM, et al. 2021. Exome and genome sequencing for pediatric patients with congenital anomalies or intellectual disability: an evidence-based clinical guideline of the American College of Medical Genetics and Genomics (ACMG). Genet Med [DOI] [PubMed] [Google Scholar]
- 57.Margulies M, Egholm M, Altman WE, Attiya S, Bader JS, et al. 2005. Genome sequencing in microfabricated high-density picolitre reactors. Nature 437: 376–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Maron JL, Kingsmore SF, Wigby K, Chowdhury S, Dimmock D, et al. 2021. Novel Variant Findings and Challenges Associated With the Clinical Integration of Genomic Testing: An Interim Report of the Genomic Medicine for Ill Neonates and Infants (GEMINI) Study. JAMA Pediatr 175: e205906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.McCandless SE, Brunger JW, Cassidy SB. 2004. The burden of genetic disease on inpatient care in a children’s hospital. Am J Hum Genet 74: 121–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.McGinnis JM, Fineberg HV, Dzau VJ. 2021. Advancing the Learning Health System. N Engl J Med 385: 1–5 [DOI] [PubMed] [Google Scholar]
- 61.McInnes G, Sharo AG, Koleske ML, Brown JEH, Norstad M, et al. 2021. Opportunities and challenges for the computational interpretation of rare variation in clinically important genes. Am J Hum Genet 108: 535–48 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Medicine Io. 2015. Genomics-Enabled Learning Health Care Systems: Gathering and Using Genomic Information to Improve Patient Care and Research: Workshop Summary (2015). Washington, D.C.: The National Academies Press. 116 pp. [PubMed] [Google Scholar]
- 63.Meng L, Pammi M, Saronwala A, Magoulas P, Ghazi AR, et al. 2017. Use of Exome Sequencing for Infants in Intensive Care Units: Ascertainment of Severe Single-Gene Disorders and Effect on Medical Management. JAMA Pediatr 171: e173438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Mestek-Boukhibar L, Clement E, Jones WD, Drury S, Ocaka L, et al. 2018. Rapid Paediatric Sequencing (RaPS): comprehensive real-life workflow for rapid diagnosis of critically ill children. J Med Genet 55: 721–28 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Miller DE, Sulovari A, Wang T, Loucks H, Hoekzema K, et al. 2021. Targeted long-read sequencing identifies missing disease-causing variation. Am J Hum Genet 108: 1436–49 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Miller DT, Lee K, Chung WK, Gordon AS, Herman GE, et al. 2021. ACMG SF v3.0 list for reporting of secondary findings in clinical exome and genome sequencing: a policy statement of the American College of Medical Genetics and Genomics (ACMG). Genet Med [DOI] [PubMed] [Google Scholar]
- 67.Miller DT, Lee K, Gordon AS, Amendola LM, Adelman K, et al. 2021. Recommendations for reporting of secondary findings in clinical exome and genome sequencing, 2021 update: a policy statement of the American College of Medical Genetics and Genomics (ACMG). Genet Med [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Mitani T, Isikay S, Gezdirici A, Gulec EY, Punetha J, et al. 2021. High prevalence of multilocus pathogenic variation in neurodevelopmental disorders in the Turkish population. Am J Hum Genet 108: 1981–2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Monaghan KG, Leach NT, Pekarek D, Prasad P, Rose NC, et al. 2020. The use of fetal exome sequencing in prenatal diagnosis: a points to consider document of the American College of Medical Genetics and Genomics (ACMG). Genet Med 22: 675–80 [DOI] [PubMed] [Google Scholar]
- 70.Ng SB, Buckingham KJ, Lee C, Bigham AW, Tabor HK, et al. 2010. Exome sequencing identifies the cause of a mendelian disorder. Nat Genet 42: 30–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Ng SB, Turner EH, Robertson PD, Flygare SD, Bigham AW, et al. 2009. Targeted capture and massively parallel sequencing of 12 human exomes. Nature 461: 272–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.OMIM. 2021. OMIM Morbid Map Scorecard. In OMIM Gene Map Statistics, ed. OMIM. OMIM: OMIM [Google Scholar]
- 73.Ouyang X, Zhang Y, Zhang L, Luo J, Zhang T, et al. 2021. Clinical Utility of Rapid Exome Sequencing Combined With Mitochondrial DNA Sequencing in Critically Ill Pediatric Patients With Suspected Genetic Disorders. Front Genet 12: 725259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Owen MJ. 2022. Genome-to-Treatment: A virtual, automated system for population-scale diagnosis and acute management guidance for genetic diseases in 13.5 hours. Nat Commun in press [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Owen MJ, Niemi AK, Dimmock DP, Speziale M, Nespeca M, et al. 2021. Rapid Sequencing-Based Diagnosis of Thiamine Metabolism Dysfunction Syndrome. N Engl J Med 384: 2159–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Perrier S, Gauquelin L, Wambach JA, Bernard G. 2021. Distinguishing severe phenotypes associated with pathogenic variants in POLR3A. Am J Med Genet A [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Petrikin JE, Cakici JA, Clark MM, Willig LK, Sweeney NM, et al. 2018. The NSIGHT1-randomized controlled trial: rapid whole-genome sequencing for accelerated etiologic diagnosis in critically ill infants. NPJ Genom Med 3: 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Platt R, Simon GE, Hernandez AF. 2021. Is Learning Worth the Trouble? - Improving Health Care System Participation in Embedded Research. N Engl J Med 385: 5–7 [DOI] [PubMed] [Google Scholar]
- 79.Powis Z, Farwell Hagman KD, Blanco K, Au M, Graham JM, et al. 2020. When moments matter: Finding answers with rapid exome sequencing. Mol Genet Genomic Med 8: e1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Ruppel H, Liu V. 2019. To catch a killer: electronic sepsis alert tools reaching a fever pitch? BMJ Qual Saf 28: 693–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Sanford E, Jones MC, Brigger M, Hammer M, Giudugli L, et al. 2020. Postmortem diagnosis of PPA2-associated sudden cardiac death from dried blood spot in a neonate presenting with vocal cord paralysis. Cold Spring Harb Mol Case Stud 6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Sanford EF, Clark MM, Farnaes L, Williams MR, Perry JC, et al. 2019. Rapid Whole Genome Sequencing Has Clinical Utility in Children in the PICU. Pediatr Crit Care Med 20: 1007–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Saunders CJ, Miller NA, Soden SE, Dinwiddie DL, Noll A, et al. 2012. Rapid whole-genome sequencing for genetic disease diagnosis in neonatal intensive care units. Sci Transl Med 4: 154ra35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Scholz T, Blohm ME, Kortum F, Bierhals T, Lessel D, et al. 2021. Whole-Exome Sequencing in Critically Ill Neonates and Infants: Diagnostic Yield and Predictability of Monogenic Diagnosis. Neonatology 118: 454–61 [DOI] [PubMed] [Google Scholar]
- 85.Shafin K, Pesout T, Lorig-Roach R, Haukness M, Olsen HE, et al. 2020. Nanopore sequencing and the Shasta toolkit enable efficient de novo assembly of eleven human genomes. Nat Biotechnol 38: 1044–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Smigiel R, Biela M, Szmyd K, Bloch M, Szmida E, et al. 2020. Rapid Whole-Exome Sequencing as a Diagnostic Tool in a Neonatal/Pediatric Intensive Care Unit. J Clin Med 9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Smith HS, Swint JM, Lalani SR, de Oliveira Otto MC, Yamal JM, et al. 2020. Exome sequencing compared with standard genetic tests for critically ill infants with suspected genetic conditions. Genet Med 22: 1303–10 [DOI] [PubMed] [Google Scholar]
- 88.Sobesky R, Guillaud O, Bouzbib C, Sogni P, Poujois A, et al. 2021. Non-invasive diagnosis and follow-up of rare genetic liver diseases. Clin Res Hepatol Gastroenterol 46: 101768. [DOI] [PubMed] [Google Scholar]
- 89.Stark Z, Dolman L, Manolio TA, Ozenberger B, Hill SL, et al. 2019. Integrating Genomics into Healthcare: A Global Responsibility. Am J Hum Genet 104: 13–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Stark Z, Lunke S, Brett GR, Tan NB, Stapleton R, et al. 2018. Meeting the challenges of implementing rapid genomic testing in acute pediatric care. Genet Med 20: 1554–63 [DOI] [PubMed] [Google Scholar]
- 91.Swaggart KA, Swarr DT, Tolusso LK, He H, Dawson DB, Suhrie KR. 2019. Making a Genetic Diagnosis in a Level IV Neonatal Intensive Care Unit Population: Who, When, How, and at What Cost? J Pediatr 213: 211–17 e4 [DOI] [PubMed] [Google Scholar]
- 92.Thakur A, Parvez MM, Leeder JS, Prasad B. 2021. Ontogeny of Drug-Metabolizing Enzymes. Methods Mol Biol 2342: 551–93 [DOI] [PubMed] [Google Scholar]
- 93.Thomas RH, Berkovic SF. 2014. The hidden genetics of epilepsy-a clinically important new paradigm. Nat Rev Neurol 10: 283–92 [DOI] [PubMed] [Google Scholar]
- 94.Tolusso LK, Hazelton P, Wong B, Swarr DT. 2021. Beyond diagnostic yield: prenatal exome sequencing results in maternal, neonatal, and familial clinical management changes. Genet Med 23: 909–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Vale AM, Schroeder HW Jr. 2010. Clinical consequences of defects in B-cell development. J Allergy Clin Immunol 125: 778–87 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.van Diemen CC, Kerstjens-Frederikse WS, Bergman KA, de Koning TJ, Sikkema-Raddatz B, et al. 2017. Rapid Targeted Genomics in Critically Ill Newborns. Pediatrics 140 [DOI] [PubMed] [Google Scholar]
- 97.Wang H, Lu Y, Dong X, Lu G, Cheng G, et al. 2020. Optimized trio genome sequencing (OTGS) as a first-tier genetic test in critically ill infants: practice in China. Hum Genet 139: 473–82 [DOI] [PubMed] [Google Scholar]
- 98.Wang H, Qian Y, Lu Y, Qin Q, Lu G, et al. 2020. Clinical utility of 24-h rapid trio-exome sequencing for critically ill infants. NPJ Genom Med 5: 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Wapner RJ. 2021. Expanding our understanding of fetal genetic diseases: the beginning of in utero precision medicine. BJOG 128: 430. [DOI] [PubMed] [Google Scholar]
- 100.Warnat-Herresthal S, Schultze H, Shastry KL, Manamohan S, Mukherjee S, et al. 2021. Swarm Learning for decentralized and confidential clinical machine learning. Nature 594: 265–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Williams JK, Cashion AK, Shekar S, Ginsburg GS. 2016. Genomics, clinical research, and learning health care systems: Strategies to improve patient care. Nurs Outlook 64: 225–8 [DOI] [PubMed] [Google Scholar]
- 102.Williams MS. 2019. Early Lessons from the Implementation of Genomic Medicine Programs. Annu Rev Genomics Hum Genet 20: 389–411 [DOI] [PubMed] [Google Scholar]
- 103.Willig LK, Petrikin JE, Smith LD, Saunders CJ, Thiffault I, et al. 2015. Whole-genome sequencing for identification of Mendelian disorders in critically ill infants: a retrospective analysis of diagnostic and clinical findings. Lancet Respir Med 3: 377–87 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Wojcik MH, Reimers R, Poorvu T, Agrawal PB. 2020. Genetic diagnosis in the fetus. J Perinatol 40: 997–1006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Wojcik MH, Schwartz TS, Thiele KE, Paterson H, Stadelmaier R, et al. 2019. Infant mortality: the contribution of genetic disorders. J Perinatol 39: 1611–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Wojcik MH, Schwartz TS, Yamin I, Edward HL, Genetti CA, et al. 2018. Genetic disorders and mortality in infancy and early childhood: delayed diagnoses and missed opportunities. Genet Med 20: 1396–404 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Worthey EA, Mayer AN, Syverson GD, Helbling D, Bonacci BB, et al. 2011. Making a definitive diagnosis: successful clinical application of whole exome sequencing in a child with intractable inflammatory bowel disease. Genet Med 13: 255–62 [DOI] [PubMed] [Google Scholar]
- 108.Wouters RHP, van der Graaf R, Rigter T, Bunnik EM, Ploem MC, et al. 2021. Towards a Responsible Transition to Learning Healthcare Systems in Precision Medicine: Ethical Points to Consider. J Pers Med 11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Wu B, Kang W, Wang Y, Zhuang D, Chen L, et al. 2021. Application of Full-Spectrum Rapid Clinical Genome Sequencing Improves Diagnostic Rate and Clinical Outcomes in Critically Ill Infants in the China Neonatal Genomes Project. Crit Care Med 49: 1674–83 [DOI] [PubMed] [Google Scholar]
- 110.Wu ET, Hwu WL, Chien YH, Hsu C, Chen TF, et al. 2019. Critical Trio Exome Benefits In-Time Decision-Making for Pediatric Patients With Severe Illnesses. Pediatr Crit Care Med 20: 1021–26 [DOI] [PubMed] [Google Scholar]