REVIEW ARTICLE


Next-generation Sequencing in Newborn Screening: A Review on Clinical and Economic Prospects



Amit K. Mittal1, *, Dolat S. Shekhawat2, 3, Vibha Joshi1, Pratibha Singh4, and, Kuldeep Singh1, 2, 3, #
1 Resource Centre Health Technology Assessment, All India Institute of Medical Sciences, Jodhpur, Rajasthan
2 NIDAN Kendra, All India Institute of Medical Sciences, Jodhpur, Rajasthan
3 Department of Paediatrics, All India Institute of Medical Sciences, Jodhpur, Rajasthan
4 Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, Jodhpur, India


Article Metrics

CrossRef Citations:
0
Total Statistics:

Full-Text HTML Views: 2562
Abstract HTML Views: 728
PDF Downloads: 669
ePub Downloads: 313
Total Views/Downloads: 4272
Unique Statistics:

Full-Text HTML Views: 1297
Abstract HTML Views: 380
PDF Downloads: 448
ePub Downloads: 234
Total Views/Downloads: 2359



Creative Commons License
© 2023 Mittal et al.

open-access license: This is an open access article distributed under the terms of the Creative Commons Attribution 4.0 International Public License (CC-BY 4.0), a copy of which is available at: https://creativecommons.org/licenses/by/4.0/legalcode. This license permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

* Address correspondence to these authors at the Health Technology Assessment Resource Centre, All India Institute of Medical Sciences Jodhpur, Basni-2, Jodhpur, Rajasthan, 342005, India; and Department of Pediatrics, All India Institute of Medical Sciences Jodhpur, Basni-2, Jodhpur, Rajasthan, 342005, India; E-mails: Kulpra@gmail.com, amitkrbiotech@gmail.com


Abstract

Next-generation sequencing (NGS) technology has revamped the area of genetics with improve the sequence of a substantial number of genes with high accuracy and short turn-around time. It may allow being used of NGS as a first-tier diagnostic test for inborn errors of metabolic or other genetic disorders. Early diagnosis of genetic disorders may help to improve the clinical condition of the child. The advantages of NGS included panel-specific gene sequencing, which targets disease-specific genes to confirm the genetic conditions. This review discussed the advantage and potential challenges of the NGS in newborn screening, other methodologies for newborn screening v/s NGS. The application of NGS in various disorders, in comparison to the clinical importance, and economic aspects of other existing technologies are also discussed. Gene-specific panels and whole exome sequencing have shortened the clinical diagnosis of complex medical conditions at an early age. Furthermore, gene sequencing facilitates to recognize the novel mutations. There are innumerable gaps in between knowledge, as well as the views of varied populations, abilities of public health, and health economics. DNA sequencing through NGS is nowadays frequently used in some clinical diagnoses, and its execution in newborn screening can provide us with better outcomes. Although inferences across the countries additional rigorous cost-effectiveness studies towards NGS have to be piloted and it is a favour to use NGS for newborn screening. In conclusion, NGS is a rapid, robust, and accurate diagnostic tool that can be used for newborn screening which helps the clinician to make a correct diagnosis and help in prior prevention and surveillance of disorder conditions.

Keywords: Newborn screening, Metabolic disorders, Next-generation sequencing, Pediatrics, Rare disorders, Economic evaluation.



1. INTRODUCTION

Next-generation sequencing (NGS) has the potential to improve the diagnostic and prognostic success of newborn screening (NBS) programs [1]. It is an effective tool for identifying various genetic alterations in newborns. NGS has transformed the field of genetics, providing the ability to sequence a substantial number of genes within a short turnaround time in a multiplexing manner [2]. The utility of NGS has dramatically increased, which may be suggestive of the particular disorder with noteworthy cost decreases and wider community acceptance [3]. As a direct result, the ability to make inherited disorders diagnoses has increased via NGS. It can be performed using dried blood spot (DBS) of neonates through isolation of DNA and has the prospective to improve the investigative and prognostic efficiency of NBS programs [4]. However, a few more demanding clinical-effectiveness studies related to NGS have to be conducted on NBS, which are required to define whether NGS can increase screening results in terms of sensitivity and specificity [5]. Several government health organizations were recognized to confirm that NBS should be standardized and uniform [6]. Since then, NBS has continued to explore the best framework moving forward as technology diagnosing children with genetic conditions continues to emerge [7]. Furthermore, there are various gaps in between knowledge, as well as the views of varied populations, abilities of public health, and clinical effectiveness inferences [8]. Keeping the following view, this article with highlights the current advancements in the usage of NGS in NBS, the obstructions in its application in NBS, and the extensive impacts of its use in the NBS program. The main objective of NGS in NBS is to be the targeted investigation and distinction of gene variants considering a suspect of preventable or curable conditions [9]. Fig. (1) summarizes the brief overview of NBS through NGS, NBS challenges, and opportunities for population health. In addition, this article provide a brief history and outline of the current state of NGS regarding NBS including recent recommendations and prospective tasks for the usage of such tools in NBS.

2. NEWBORN SCREENING FOR VARIOUS GENETIC DISORDERS

NBS program was introduced by the World Health Organization (WHO) as per Wilson and Jungner guidelines for the development and implementation of a screening test [10]. Although the tests supplementary provided the outline for the additional development and amendment of NBS [11]. It is a public health program, which aims to identify pediatric populations with medically unfit conditions earlier than becoming symptomatic [12]. Over the past few decades, the number of medical conditions in children has gradually increased. These medical conditions are auxiliary to the screening program in different areas across the world [13]. The NBS program interventions can provide long-term results in terms of improvement and carrier identification [14]. The program was underway with identifying phenylketonuria (PKU), hemoglobinopathies to a diagnosis of sickle cell disease, glucose-6-phosphate dehydrogenase (G6PD) deficiency, congenital hypothyroidism (CH), congenital adrenalhyperplasia (CAH), tyrosinemia type I (HT1), cystic fibrosis (CF), etc [15]. The diagnosis and exploration of these disorders are challenging and time-consuming due to the rarity of the individual disorder, the genetic heterogeneity, and variability of the clinical manifestations, and the lack of laboratory testing [16]. Various NBS programs have been experienced in frequent countries since the start of NBS [17]. NBS was started in the 1960s and phenylketonuria screening was successfully implemented by different nations across the world first [12].

3. NEWER TECHNIQUES TO EXPAND NEWBORN SCREENING PROGRAM

Several health organizations were recognized to confirm that NBS should be standardized and uniform. Since, the field of NBS has continued to explore the best framework moving forward as technology for diagnosing pediatric populations with genetic conditions continues to emerge [8, 18, 19]. With the progress and the availability of electrospray ionization (ESI) tandem mass spectrometry (MS/MS), the instrument can measure various metabolites related to inborn errors of metabolism (IEM) disorders [20]. It was the first great extension of the NBS programs across the world. Furthermore, future development for the screening of rare disorders also needs to be required, which needs additional diagnostic tests [21]. To facilitate more accuracy, a combined screening test can be used MS/MS and targeted-NGS in a parallel way, which may be more useful to offer the potential to enhance performance [22]. It can increase the specificity results by identifying new disease-associated variants along with specifying genotypes to identify the carriers [23]. Fig. (2) summarizes the intermittent techniques used individually or combinedly for the NBS.

Fig. (1). Schematic view of the newborn screening procedure through next-generation sequencing, opportunities, and challenges from a public health perspective.

Fig. (2). Various newborn screening methods/techniques are used in the NBS program to diagnose developmental, genetic, and metabolic disorders.

4. NEXT-GENERATION SEQUENCING IN NEWBORN SCREENING PROGRAM

NGS has cutting-edge technology and transformed the field of genetics using molecular biology, providing the ability to sequence a large number of important genes within a short turnaround time [24]. As a direct result, the ability to make genetic diagnoses has increased powerfully [25]. Genome sequencing is now often used in various medical diagnoses, and its implementation in NBS has also been extensively encouraged [26]. In the last decade, there is a significant advancement toward high-throughput DNA sequencing, which resulted in a reduction in both the cost and time required for genetic analysis [1, 9]. These methods are widely used in massive multi-parallel sequencing. The NGS can investigate all protein-coding genes or even entire genomes in one single time via whole-exome sequencing (WES) or whole-genome sequencing (WGS) [27]. The ease of accessibility and a considerable decrease in costs of DNA sequencing has raised the ultimatum of whether metabolic NBS should be replaced by WES/WGS [8]. While NGS can potentially identify the number of disorders and the generalization of its practice raises many important ethical issues [28]. These developments can result in an escalation of genetic diagnoses in a shorter time [29]. Fig. (3) summarizes the common recommended diseases for NBS and their targeted genes.

Despite the health benefit of genetic testing in the clinical setting, the testing in asymptomatic children has been widely questioned [30]. Given the limited knowledge of rare genetic conditions, large prospective studies of healthy individuals should be incorporated in the decision to include conditions on an NGS-NBS panel, especially in circumstances with no current actionable treatment [26]. Genomic information can help manage health throughout life and can be applied to thoughtful children [23, 31]. Nevertheless, the current research focusing on long-term outcomes and psychological impact is needed before applying the technology to population screening [32]. Milko et al. piloted the NBS through NGS, and the Newborn Exome Sequencing for Universal Screening (NC NEXUS) was conducted to assess the technical feasibility and limitations of NGS-NBS. Furthermore, it was planned and evaluated framework to convey various types of genetic information [33]. Recently, NeoSeq, an economic genomic screening test for NBS utilized for the most inborn errors of metabolism, which can reduce the rate of false positive results, shorten the porting cycles, and reduce the screening cost also [22, 34]. Genetic disorders are the foremost reason for mortality and morbidity in newborns in neonatal and pediatric intensive care units (NICU/PICU) [35]. One study reported that rapid whole-genome sequencing (rWGS) increased the proportion of NICU/PICU infants receiving a genetic diagnosis within 28 days [36]. In China, Children’s hospitals developed a panel of 465 causative genes for 596 early-onset, relatively high incidence, and potentially actionable severe inherited diseases in NBS with the Targeted Sequencing (NESTS) program and suggest that NESTS is feasible and cost-effective as a first-tier NBS program, which can change the status of current clinical practice of NBS in China [37]. However, it is still necessary to further optimize the panel design and add more clinically relevant genomic variants to increase its sensiti-

Fig. (3). Core recommended disease for the newborn screening program. The classification of the type of disorders, disease name, and their targeted genes are described.

vity [22]. Boemer et al. 2017 reported that NGS using dried blood spots can detect causal mutations in patients with inherited metabolic disorders [38]. The NGS-based NBS approach is idyllic for the identification of newborns with monogenic forms of IEM, which are heterogeneous but can be easily distinguished [39]. The NGS with the amplicon-based panel has also been validated for diagnostic purposes and can be used for second-tier NBS IEM tests [40].

5. NEXT-GENERATION SEQUENCING ON THE WAY TO CLINICAL AND ECONOMIC VIEWPOINTS IN THE NEWBORN SCREENING PROGRAM

The extension of NBS has raised many clinical, ethical, lawful, psychological, sociological, and scientific distresses over time [1]. As the cost of NGS continues to decrease and our understanding of genomic databases continues to improve, demands have been elevated to apply NGS on a larger scale with improved outcomes for potential health concerns [41]. There are various gaps in between facts, as well as the views of diverse populations, capabilities of health care systems, and health economic implications. It will be essential to scrupulously evaluate the outcomes and ensure the plans can evolve to maximize benefit. The ease of accessibility and substantial decrease in costs of DNA sequencing has raised the question of whether metabolic NBS should be replaced by WGS/WES [42]. While in newborn genome sequencing can potentially increase the number of disorders identified and the simplification of its practice raises some important ethical issues [28]. The development of the map of the human genome assured a new era in medicine [43]. In the last few decades, the usefulness of DNA sequencing for health benefits has been demonstrated during manifold periods and is now a routine in some areas of medicine [43]. Advanced DNA sequencing techniques and truncated cost offered the promise of a personalized genome to provide a personal health benefit [44]. The first usage of genome sequencing for disease diagnosis befell in 2009 at the University of Wisconsin for a child with severe inflammatory bowel disease. The cause of the disease is the occurrence of the pathogenic variant of the XIAP gene [45]. The newborn screening cost via the traditional method in Europe is 1 to 44 Euro (€1: screening for 2 conditions; €43.24: screening for 17 conditions) and in the United States is around 100 USD ($100: screening for 30 conditions) per test [1]. Few studies should be commenced to obtain accurate data about all costs involved as part of NBS including subsequent diagnostic costs as well as the costs of not implementing NGS. The price of NGS for million bases (i.e. 1 Mb) has considerably fallen and may continue to fall even extra. However, the cost of NGS is still high other than the existing NBS programs. The cost of NGS in NBS, pre-and post-testing, is also a key issue in the decision to implement NGS in NBS. Furthermore, the setup and human resource costs to ensure monitoring, appropriate education, counselling, interventions, and storage needs to be assessed [9, 30].

CONCLUSION AND FUTURE PROSPECTS

NGS is a rapid, robust, and accurate diagnostic tool that can be used for newborn screening, which helps the clinician to make a correct diagnosis and help in prior prevention and surveillance of disorder conditions. It is of substantial interest whether NGS is applicable for NBS and has great potential to improve NBS through the understanding of genetics, which can provide flawless diagnosis. The expansion of the NBS program for various disorders needs to be required, which calls for additional tests. Necessary characteristics of a screening test include the availability of an economical method of identification of important health problems for which treatment is available, as well as the infrastructure to provide such treatment. It can sequence all protein-coding regions or even entire genomes at one single time, resulting in an increase in genetic diagnoses and a shortened time to diagnosis for patients with expected genetic disorders. A broader understanding of several aspects is needed to facilitate the incorporation of NGS into newborn public health screening programs and inform the development of important future NBS policy guidelines.

LIST OF ABBREVIATIONS

DBS = Dried blood spot
IEM = Inborn errors of metabolism
MS/MS = Tandem mass spectrometry
NBS = Newborn screening
NGS = Next-generation sequencing
WES = Whole exome sequencing
WGS = Whole genome sequencing

CONSENT FOR PUBLICATION

Not applicable.

FUNDING

None.

CONFLICT OF INTEREST

Dr. Amit Kumar Mittal is an Editorial Advisory Board of The Open Biotechnology Journal.

ACKNOWLEDGEMENTS

The authors AKM and VJ would like to thank the Department of Health Research (DHR) and author DSS would like to thank the UMMID initiative, the Department of Biotechnology (DBT), Govt. of India for the financial support.

REFERENCES

[1] Remec ZI, Trebusak Podkrajsek K, Repic Lampret B, et al. Next-generation sequencing in newborn screening: A review of current state. Front Genet 2021; 12: 662254.
[2] Kulski JK. Next-Generation Sequencing — An Overview of the History, Tools, and “Omic” Applications. In: Next Generation Sequencing - Advances, Applications and Challenges (pp 3-60) (Biochemistry, Genetics and Molecular Biology). InTech 2016.
[3] Tan O, Shrestha R, Cunich M, Schofield DJ. Application of next-generation sequencing to improve cancer management: A review of the clinical effectiveness and cost-effectiveness. Clin Genet 2018; 93(3): 533.
[4] Lefterova MI, Shen P, Odegaard JI, et al. Next-generation molecular testing of newborn dried blood spots for cystic fibrosis. J Mol Diagn 2016; 18(2): 267-82.
[5] van Campen JC, Sollars ESA, Thomas RC, et al. Next generation sequencing in newborn screening in the United Kingdom National Health Service. Int J Neonatal Screen 2019; 5(4): 40.
[6] Blom M, Zetterström RH, Stray-Pedersen A, et al. Recommendations for uniform definitions used in newborn screening for severe combined immunodeficiency. J Allergy Clin Immunol 2022; 149(4): 1428-36.
[7] Phornphutkul C, Padbury J. Large scale next generation sequencing and newborn screening: Are we ready? J Pediatr 2019; 209: 9-10.
[8] Woerner AC, Gallagher RC, Vockley J, Adhikari AN. The use of whole genome and exome sequencing for newborn screening: Challenges and opportunities for population health. Front Pediatr 2021; 9: 663752.
[9] Howard HC, Knoppers BM, Cornel MC, Wright Clayton E, Sénécal K, Borry P. Whole-genome sequencing in newborn screening? A statement on the continued importance of targeted approaches in newborn screening programmes. Eur J Hum Genet 2015; 23(12): 1593-600.
[10] World Health Organization. Screening programmes: A short guide. Increase effectiveness, maximize benefits and minimize harm. Available from: apps.who.int
[11] Kanungo S, Patel DR, Neelakantan M, Ryali B. Newborn screening and changing face of inborn errors of metabolism in the United States. Ann Transl Med 2018; 6(24): 468.
[12] Pitt JJ. Newborn screening. Clin Biochem Rev 2010; 31(2): 57-68.
[13] Koracin V, Mlinaric M, Baric I, et al. Current status of newborn screening in Southeastern Europe. Front Pediatr 2021; 9: 648939.
[14] Cornel MC, Rigter T, Jansen ME, Henneman L. Neonatal and carrier screening for rare diseases: How innovation challenges screening criteria worldwide. J Community Genet 2021; 12(2): 257-65.
[15] Knapkova M, Hall K, Loeber G, et al. Reliability of neonatal screening results. Int J Neonatal Screen 2018; 4(3): 28.
[16] Liu Z, Zhu L, Roberts R, Tong W. Toward clinical implementation of next-generation sequencing-based genetic testing in rare diseases: Where are we? Trends Genet 2019; 35(11): 852-67.
[17] Therrell BL Jr, Padilla CD. Newborn screening in the developing countries. Curr Opin Pediatr 2018; 30(6): 734-9.
[18] Abhyankar S, Lloyd-Puryear MA, Goodwin R, et al. Standardizing newborn screening results for health information exchange. AMIA Annu Symp Proc 2010; 2010: 1-5.
[19] Kapoor S, Kabra M. Newborn screening in India: Current perspectives. Indian Pediatr 2010; 47(3): 219-24.
[20] Lim JS, Tan ES, John CM, et al. Inborn Error of Metabolism (IEM) screening in Singapore by electrospray ionization-tandem mass spectrometry (ESI/MS/MS): An 8year journey from pilot to current program. Mol Genet Metab 2014; 113(1-2): 53-61.
[21] Kapoor S, Thelma BK. Status of newborn screening and inborn errors of metabolism in India. Indian J Pediatr 2018; 85(12): 1110-7.
[22] Wang H, Yang Y, Zhou L, Wang Y, Long W, Yu B. NeoSeq: A new method of genomic sequencing for newborn screening. Orphanet J Rare Dis 2021; 16(1): 481.
[23] Johnston J, Lantos JD, Goldenberg A, Chen F, Parens E, Koenig BA. Sequencing newborns: A call for nuanced use of genomic technologies. Hastings Cent Rep 2018; 48(Suppl. 2): S2-6.
[24] Dahui Q. Next-generation sequencing and its clinical application. Cancer Biol Med 2019; 16(1): 4-10.
[25] Adams DR, Eng CM. Next-generation sequencing to diagnose suspected genetic disorders. N Engl J Med 2018; 379(14): 1353-62.
[26] Berg JS, Agrawal PB, Bailey DB Jr, et al. Newborn sequencing in genomic medicine and public health. Pediatrics 2017; 139(2): e20162252.
[27] Smith LD, Willig LK, Kingsmore SF. Whole-exome sequencing and whole-genome sequencing in critically ill neonates suspected to have single-gene disorders. Cold Spring Harb Perspect Med 2016; 6(2): a023168.
[28] Reinstein E. Challenges of using next generation sequencing in newborn screening. Genet Res 2015; 97: e21.
[29] Green NS, Dolan SM, Murray TH. Newborn screening: Complexities in universal genetic testing. Am J Public Health 2006; 96(11): 1955-9.
[30] Kelly N, Makarem DC, Wasserstein MP. Screening of newborns for disorders with high benefit-risk ratios should be mandatory. J Law Med Ethics 2016; 44(2): 231-40.
[31] Bilkey GA, Burns BL, Coles EP, et al. Genomic testing for human health and disease across the life cycle: applications and ethical, legal, and social challenges. Front Public Health 2019; 7: 40.
[32] Jalan AB, Kudalkar KV. Newborn screening: Need of the hour. Karnataka Pediatric Journal 2021; 36(1): 35-41.
[33] Milko LV, Rini C, Lewis MA, et al. Evaluating parents’ decisions about next-generation sequencing for their child in the NC NEXUS (North Carolina Newborn Exome Sequencing for Universal Screening) study: A randomized controlled trial protocol. Trials 2018; 19(1): 344.
[34] Wu X, Yang Y, Zhou L, Long W, Yu B. Are we ready for newborn genetic screening? A cross-sectional survey of healthcare professionals in Southeast China. Front Pediatr 2022; 10: 875229.
[35] Farnaes L, Hildreth A, Sweeney NM, et al. Rapid whole-genome sequencing decreases infant morbidity and cost of hospitalization. NPJ Genom Med 2018; 3(1): 10.
[36] Petrikin JE, Cakici JA, Clark MM, et al. The NSIGHT1-randomized controlled trial: rapid whole-genome sequencing for accelerated etiologic diagnosis in critically ill infants. NPJ Genom Med 2018; 3(1): 6.
[37] Hao C, Guo R, Hu X, et al. Newborn screening with targeted sequencing: A multicenter investigation and a pilot clinical study in China. J Genet Genomics 2022; 49(1): 13-9.
[38] Boemer F, Fasquelle C, d’Otreppe S, et al. A next-generation newborn screening pilot study: NGS on dried blood spots detects causal mutations in patients with inherited metabolic diseases. Sci Rep 2017; 7(1): 17641.
[39] Pourfarzam M, Zadhoush F. Newborn screening for inherited metabolic disorders; news and views. J Res Med Sci 2013; 18(9): 801.
[40] Tsang KY, Chan TCH, Yeung MCW, Wong TK, Lau WT, Mak CM. Validation of amplicon-based next generation sequencing panel for second-tier test in newborn screening for inborn errors of metabolism. J Lab Med 2021; 45(6): 267-74.
[41] McCombie WR, McPherson JD. Future promises and concerns of ubiquitous next-generation sequencing. Cold Spring Harb Perspect Med 2019; 9(9): a025783.
[42] Mujamammi AH. Insights into National laboratory newborn screening and future prospects. Medicina 2022; 58(2): 272.
[43] Trier C, Fournous G, Strand JM, Stray-Pedersen A, Pettersen RD, Rowe AD. Next-generation sequencing of newborn screening genes: the accuracy of short-read mapping. NPJ Genom Med 2020; 5(1): 36.
[44] Phillips KA, Trosman JR, Kelley RK, Pletcher MJ, Douglas MP, Weldon CB. Genomic sequencing: assessing the health care system, policy, and big-data implications. Health Aff 2014; 33(7): 1246-53.
[45] Worthey EA, Mayer AN, Syverson GD, et al. Making a definitive diagnosis: Successful clinical application of whole exome sequencing in a child with intractable inflammatory bowel disease. Genet Med 2011; 13(3): 255-62.