Hostname: page-component-848d4c4894-m9kch Total loading time: 0 Render date: 2024-05-16T19:13:32.749Z Has data issue: false hasContentIssue false

Precision diagnostics in children

Published online by Cambridge University Press:  03 February 2023

Paul Dimitri*
Affiliation:
Department of Paediatric Endocrinology, Sheffield Children’s NHS Foundation Trust, Sheffield, UK The College of Health, Wellbeing and Life Sciences, Sheffield Hallam University, Sheffield, UK
*
Author for correspondence: Paul Dimitri, Email: paul.dimitri@nhs.net
Rights & Permissions [Opens in a new window]

Abstract

Medical practice is transforming from a reactive to a pro-active and preventive discipline that is underpinned by precision medicine. The advances in technologies in such fields as genomics, proteomics, metabolomics, transcriptomics and artificial intelligence have resulted in a paradigm shift in our understanding of specific diseases in childhood, greatly enhanced by our ability to combine data from changes within cells to the impact of environmental and population changes. Diseases in children have been reclassified as we understand more about their genomic origin and their evolution. Genomic discoveries, additional ‘omics’ data and advances such as optical genome mapping have driven rapid improvements in the precision and speed of diagnoses of diseases in children and are now being incorporated into newborn screening, have improved targeted therapies in childhood and have supported the development of predictive biomarkers to assess therapeutic impact and determine prognosis in congenital and acquired diseases of childhood. New medical device technologies are facilitating data capture at a population level to support higher diagnostic accuracy and tailored therapies in children according to predicted population outcome, and digital ecosystems now tailor therapies and provide support for their specific needs. By capturing biological and environmental data as early as possible in childhood, we can understand factors that predict disease or maintain health and track changes across a more extensive longitudinal path. Data from multiple health and external sources over long-time periods starting from birth or even in the in utero environment will provide further clarity about how to sustain health and prevent or predict disease. In this respect, we will not only use data to diagnose disease, but precision diagnostics will aid the ‘diagnosis of good health’. The principle of ‘start early and change more’ will thus underpin the value of applying a personalised medicine approach early in life.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2023. Published by Cambridge University Press

Impact statement

There are 1.8 billion young people in the world today – 40% of the global population is under 24. We should aim to support children and young people with maintaining a long and healthy life and develop a system by which we personalise their health and healthcare for a better future. New advances in the fields of genomics, proteomics and cytogenetics have revolutionised our ability to understand the mechanisms underpinning the evolution and manifestation of existing diseases, discover new diseases, develop targeted therapies for specific populations of children to improve outcomes and predict children’s response to therapy, their risk of relapse and their prognosis. Importantly, our ability to diagnose disease early in life, and in particular using genomics in newborn screening supports early intervention and prevention and provides a greater understanding of the evolution and manifestation of diseases over longer periods. Critical to this process is the study of disease in the context of exposome, a new paradigm that encompasses the totality of human environmental exposures from conception onwards. A personalised approach to children’s health requires knowledge of the patient as an individual and their surrounding ecosystem. New technologies that can provide accurate tracking and recording of environmental data and technologies that can generate predictive models relating to future outcomes will add to the factors that facilitate a precision-based approach to managing health and disease over the life course. Furthermore, the ability to capture health data across large populations of children allows us to understand population characteristics in specific diseases that in turn drive individual interventions and changes. Precision medicine, precision diagnostics and the technological advances underpinning these rapidly advancing fields will change the way in which we understand disease, sustain health and improve quality of life. Focussing our efforts early in life is an investment for future health and healthcare.

Introduction

Healthcare is evolving from a traditional ‘one-size-fits-all’ approach to a model that accounts for the prediction of individual patient disease risks, a tailored approach to the investigation and the development of targeted interventions. A better understanding of the underlying mechanisms and causes of rare and chronic diseases combined with technological advancements are transforming medicine from a reactive to a pro-active and preventive discipline that has been encapsulated in the ‘4P’ approach to medicine (also referred to as P4 Medicine) to maintain health and well-being and to prevent and predict disease and thus respond accordingly to individual needs – ‘4P – Preventive, Predictive, Participatory and Personalised’ (Hood et al., Reference Hood, Heath, Phelps and Lin2004; Flores et al., Reference Flores, Glusman, Brogaard, Price and Hood2013). A personalised approach to medicine requires knowledge of the patient as an individual and their surrounding ecosystem, by understanding their molecular and genetic components, their cells and tissues, the whole person and the population and environment in which they exist. This breadth and depth of knowledge of an individual and their environmental interaction underpins the science of systems biology (National Research Council (US), 2009). Systems biology is founded on the principle that the interaction between individual parameters at the genetic, molecular and cellular level with social and environmental parameters as the component parts of complex biological systems supports a more ‘predictive’ approach by which computational models can be used to predict outcomes, and individuals can be stratified more comprehensively according to their disease risks. Systems medicine focuses on the approach of systems biology to human disease by combining knowledge that ranges from individual genomic sequencing to understanding global data sets that track patient populations and their interaction with the environment (Hood et al., Reference Hood, Balling and Auffray2012).

Advances in technology and computational analysis over the last 20 years have radically enhanced our ability to collect, store and analyse data to create biological networks derived from computational models that demonstrate how factors within biological systems can maintain health or lead to disease. Importantly, factors within these computational models can be modified to predict an individual’s risk of future ill-health. Ultimately, the aim of systems medicine is to develop an ‘individual data profile’ founded upon multi-dimensional longitudinal health data, ranging from the individuals genomic/proteomic profile, hospital data from historic interactions and lifestyle and environmental data. These data have two fundamental origins – data that are derived when an individual interacts with a system such as a hospital or for health research, or alternatively, when the individual offers or inputs the data as part of an interaction with a digital platform encompassed in the participatory component of the 4P model. The latter of these has been facilitated by the advance in technologies that allow individual consumers to input and track their own health data and interact with others to monitor their health and disease risk. Portable devices such as mobile phones, tablets and laptops, novel sensors and digital platforms help to facilitate the participatory component of the 4P model extending health data collection and analysis into homes, workplaces and schools. Data collection from new devices can be passive, such that the individual allows the device to track activity without direct input, or active by which the individual inputs the data. In principle, if information can be collected early in life, this has the potential to provide richer longitudinal data about health, disease, lifestyle and environmental interaction, which could help modify factors that prevent disease, support earlier diagnoses or maintain health. Returning to the systems medicine approach, data could be derived from a number of sources including primary and secondary care health data, genomics data derived from genomic newborn screening and whole genome sequencing (WGS), educational data from schools and universities, and additional data on lifestyle and environmental exposure, sometimes referred to as the exposome, encompassing environmental exposure from the prenatal period onwards. For children, the exposome encompasses general exposure to the external environment such as climate, familial and social factors and education, and specific exposure such as infections, radiation, home factors such as diet, smoking and alcohol consumption in parents, and physical activity. The challenge as we develop tools to individualise healthcare and to prevent and predict disease in children and young people is our ability to acquire data from multiple sources including those which are controlled by the consumer. Data captured from an early age will rely on empowering children, young people and their families to permit the use of their data, particularly data that may be from non-health related and personalsources, trusting that their data will be utilised appropriately to support their health and well-being and will be stored securely (National Research Council (US), 2011). The value of using data from multiple sources must be easily demonstrable to families in a way that prevention, disease modification and individualised care in the future are supported by meaningful and actionable information that is in a way that children, young people and parents can understand, and in a way that parents can utilise to support their children in preventing ill-health or seeking earlier healthcare support.

Precision diagnostics is a branch of precision medicine using the 4P approach by which individual diseases are diagnosed based upon genomic variation, lifestyle and the environment. Understanding subpopulations with similar genomic information and the environmental and lifestyle factors that predict disease onset allows healthcare professionals and individuals to prevent disease onset or intervene early to prevent disease progression. This approach has the potential to develop a radically more cost-effective approach to healthcare, particularly in the paediatric population, where rare diseases are more prevalent. Moreover, a better understanding of modifiable lifestyle factors in childhood may help to predict or prevent future disease, thus underpinning the value of prospective data collection. Tailored therapies by stratifying individuals into subgroups according to their disease profile, response to therapy and prognosis and preventing disease through risk factor modification will result in a reduction in healthcare expenditure (Wang et al., Reference Wang, Cho, Wong and Liu2017). Furthermore, understanding the molecular and cellular origins of disease in children and young people has resulted in a paradigm shift in thinking about ‘causes’ rather than ‘symptoms’ of disease leading to a proposal for a new taxonomy for disease classification that could change the approach to clinical decision-making, diagnosis, therapy and prognosis (Blower et al., Reference Blower, Swallow, Maturana, Stones, Phillips, Dimitri, Marshman, Knapp, Dean, Higgins, Kellar, Curtis, Mills and Martin-Kerry2020).

Disease reclassification and predicting future disease in children

Precision diagnostics is founded on the ability to predict disease and the ability to adopt a personalised approach. The understanding of a disease at a molecular level will determine the onset of the disease or symptoms, or when a perturbation within a biological system will result in the manifestation of the disease. This will vary between individuals, and thus, the understanding of the biological environment at a molecular level is fundamental in determining the individual’s disease susceptibility, the severity of the disease, response to specific therapies and prognosis. At a genetic level, mutations range from loss or gain of entire chromosomes, loss of smaller regions of DNA, for example, copy number variants (CNVs), to changes in the structure of the genome in the form of translocations, inversions and insertions, or changes in the sequence of the nucleotides (Lalonde et al., Reference Lalonde, Rentas, Lin, Dulik, Skraban and Spinner2020). Advances in genomics, and in particular the development of next-generation sequencing (Lalonde et al., Reference Lalonde, Rentas, Lin, Dulik, Skraban and Spinner2020), have had a transformative effect on diagnostics in children, resulting in targeted panels for specific diagnoses, and examination of the entire exome or genome based upon clinical manifestations. Whole exome sequencing and Whole Genome Sequencing (WGS) have the potential to increase the diagnostic yield by up to 50% and as such there has been a call to adopt WGS as a first-tier test, particularly conditions in children with genetic heterogeneity (Stavropoulos et al., Reference Stavropoulos, Merico, Jobling, Bowdin, Monfared, Thiruvahindrapuram, Nalpathamkalam, Pellecchia, Yuen, Szego, Hayeems, Shaul, Brudno, Girdea, Frey, Alipanahi, Ahmed, Babul-Hirji, Porras, Carter, Chad, Chaudhry, Chitayat, Doust, Cytrynbaum, Dupuis, Ejaz, Fishman, Guerin, Hashemi, Helal, Hewson, Inbar-Feigenberg, Kannu, Karp, Kim, Kronick, Liston, MacDonald, Mercimek-Mahmutoglu, Mendoza-Londono, Nasr, Nimmo, Parkinson, Quercia, Raiman, Roifman, Schulze, Shugar, Shuman, Sinajon, Siriwardena, Weksberg, Yoon, Carew, Erickson, Leach, Klein, Ray, Meyn, Scherer, Cohn and Marshall2016; Meng et al., Reference Meng, Pammi, Saronwala, Magoulas, Ghazi, Vetrini, Zhang, He, Dharmadhikari, Qu, Ward, Braxton, Narayanan, Ge, Tokita, Santiago-Sim, Dai, Chiang, Smith, Azamian, Robak, Bostwick, Schaaf, Potocki, Scaglia, Bacino, Hanchard, Wangler, Scott, Brown, Hu, Belmont, Burrage, Graham, Sutton, Craigen, Plon, Lupski, Beaudet, Gibbs, Muzny, Miller, Wang, Leduc, Xiao, Liu, Shaw, Walkiewicz, Bi, Xia, Lee, Eng, Yang and Lalani2017; Posey et al., Reference Posey, Harel, Liu, Rosenfeld, James, Coban Akdemir, Walkiewicz, Bi, Xiao, Ding, Xia, Beaudet, Muzny, Gibbs, Boerwinkle, Eng, Sutton, Shaw, Plon, Yang and Lupski2017; Lionel et al., Reference Lionel, Costain, Monfared, Walker, Reuter, Hosseini, Thiruvahindrapuram, Merico, Jobling, Nalpathamkalam, Pellecchia, Sung, Wang, Bikangaga, Boelman, Carter, Cordeiro, Cytrynbaum, Dell, Dhir, Dowling, Heon, Hewson, Hiraki, Inbar-Feigenberg, Klatt, Kronick, Laxer, Licht, MacDonald, Mercimek-Andrews, Mendoza-Londono, Piscione, Schneider, Schulze, Silverman, Siriwardena, Snead, Sondheimer, Sutherland, Vincent, Wasserman, Weksberg, Shuman, Carew, Szego, Hayeems, Basran, Stavropoulos, Ray, Bowdin, Meyn, Cohn, Scherer and Marshall2018). Many of these conditions are congenital, manifesting at birth, infancy or early childhood. A genetic diagnosis can then inform prognosis, anticipatory care and surveillance, targeted management and future family planning. Even within conditions harbouring the same ‘macro’ genetic mutation, there can be a high degree of variability based upon the location of the specific mutation and the interaction with other genes within the same molecular pathway (Costain et al., Reference Costain, Cohn and Malkin2020). In turn, this allows individualised planning, targeted therapies and risk profiling.

This approach has led to significant clarification in disorders with heterogeneous manifestations which present in childhood, and the reclassification of disease based upon genotype rather than clinical presentation. For example, the heterogeneous clinical manifestations of Noonan syndrome, a condition that presents with a distinct facial phenotype, short stature, cardiac anomalies, developmental delay and other features (Dahlgren and Noordam, Reference Dahlgren and Noordam2022), have been reclassified and incorporated into a group of genetic mutations that are collectively known as the RAS-opathies (Gripp et al., Reference Gripp, Schill, Schoyer, Stronach, Bennett, Blaser, Brown, Burdine, Burkitt-Wright, Castel, Darilek, Dias, Dyer, Ellis, Erickson, Gelb, Green, Gross, Ho, Holder, Inoue, Jelin, Kennedy, Klein, Kontaridis, Magoulas, McConnell, McCormick, Neel, Prada, Rauen, Roberts, Rodriguez-Viciana, Rosen, Rumbaugh, Sablina, Solman, Tartaglia, Thomas, Timmer, Venkatachalam, Walsh, Wolters, Yi, Zenker and Ratner2020). RAS-opathies define germline mutations in genes that encode components or regulators of the Ras/mitogen-activated protein kinase pathway (Rauen, Reference Rauen2013) and share many of the phenotypic manifestations of Noonan syndrome. RAS-opathies also include neurofibromatosis type 1, Noonan syndrome with multiple lentigines, Noonan syndrome-like disorder with loose anagen hair, Noonan syndrome-like disorder with or without juvenile myelomonocytic leukaemia, capillary malformation-arteriovenous malformation syndrome, Costello syndrome, cardio-facio-cutaneous syndrome, SYNGAP1-related intellectual disability and Legius syndrome. Mutations associated with Noonan syndrome alone include CBL, BRAF, KRAS, LZTR1, MAP2K1 (MEK1), NRAS, PTPN11, RAF1, RIT1, SOS1 and SOS2 (Riller and Rieux-Laucat, Reference Riller and Rieux-Laucat2021; Leoni et al., Reference Leoni, Blandino, Delogu, De Rosa, Onesimo, Verusio, Marino, Lanza, Rigante, Tartaglia and Zampino2022). Thus, this provides an example of one of many genetic disorders in children that may have been mistakenly classified according to facial and systemic phenotype, or previously described as ‘non-classical’ or ‘atypical’ which can now be accurately genotyped to define the appropriate disorder, the predicted consequences, therapy and prognosis.

Similarly, osteogenesis imperfecta (OI, brittle bone disease), a rare disease in childhood, was originally classified by Sillence in 1979 into four subtypes (OI I–IV) based upon mode of inheritance, clinical manifestation and severity (Sillence et al., Reference Sillence, Senn and Danks1979). However, 20 different subtypes of this condition have emerged based upon a more refined understanding of the wide range of genes that encode relevant proteins. These proteins are involved in processes such as the synthesis of type I collagen and the differentiation, regulation and activity of bone-forming cells (osteoblasts) and bone-absorbing cells (osteoclasts). Importantly, this has led to an exponential rise in targeted therapies for bone fragility in OI based upon the genotype-derived causative mechanisms, thus supporting the value of genomic reclassification and precision diagnostics to develop novel targeted therapies for rare diseases in children (Zaripova and Khusainova, Reference Zaripova and Khusainova2020). The reduction in the cost of genomic sequencing is facilitating significant advances in the classification of diseases but additionally in predicting the onset of disease. In the UK, Genomics England has embarked on a discovery science programme aiming to sequence and analyse the whole genome in up to 200,000 babies for a set of actionable genetic conditions which may affect their health in early years. This aims to ensure timely diagnosis, access to treatment pathways and enable better outcomes and quality of life for babies and their families (Genomics England, 2021). Rapid genomic testing in critically ill-children is also advancing the field of precision diagnostics and therapy in paediatrics. The first national healthcare system-funded implementation of rapid genomic sequencing for acutely unwell children commenced in England on 1st October 2019 led by the NHS Genomic Medicine Service. The diagnostic yield was 38% with the molecular diagnosis directing management in 94% of patients (Stark and Ellard, Reference Stark and Ellard2022).

Next-generation cytogenetics such as optical genome mapping (OGM) has also advanced the speed and ability of detecting genetic abnormalities (Lam et al., Reference Lam, Hastie, Lin, Ehrlich, Das, Austin, Deshpande, Cao, Nagarajan, Xiao and Kwok2012). Cytogenetics is the genetic discipline that examines chromosomes for abnormalities. Karyotyping, chromosomal microarray analysis (CMA), multiple ligation-dependent probe amplification and fluorescent in situ hybridisation (FISH) are examples of currently utilised cytogenetic techniques but have limited resolution or limited region coverage. For example, CMA, despite having higher resolution for the detection of repeat regions of the genome known as CNVs, is limited in detecting balanced translocations and inversions (Levy and Wapner, Reference Levy and Wapner2018). Next-generation sequencing methods can detect sequence variants, but they are unable to accurately resolve CNVs. Moreover, complete genetic profiling in specific diseases either for diagnosis or for risk stratification of prognosis often relies on a combination of techniques which is time consuming and costly. OGM is a novel technology that analyses ultra-high molecular weight DNA molecules that provide a high-resolution genome-wide image. Each patient’s unique map is aligned to a reference genome map to detect CNVs and structural anomalies. OGM has strong concordance with other cytogenetic techniques such as FISH and CMA but has also advanced the detection of additional chromosomal abnormalities that are clinically relevant and that are inaccessible to standard techniques, allowing for further disease stratification in diseases such as acute lymphoblastic leukaemia (ALL) (Lestringant et al., Reference Lestringant, Duployez, Penther, Luquet, Derrieux, Lutun, Preudhomme, West, Ouled-Haddou, Devoldere, Marolleau, Garçon, Jedraszak and Ferret2021; Rack et al., Reference Rack, De Bie, Ameye, Gielen, Demeyer, Cools, De Keersmaecker, Vermeesch, Maertens, Segers, Michaux and Dewaele2022). OGM is now recognised as key genomic technology capable of detecting all classes of structural variants in many disorders, which will undoubtedly improve the speed and accuracy of diagnosis of rare disorders in children (Levy and Wapner, Reference Levy and Wapner2018; Mantere et al., Reference Mantere, Neveling, Pebrel-Richard, Benoist, van der Zande, Kater-Baats, Baatout, van Beek, Yammine and Oorsprong2020) but also improve risk stratification in common haematological disorders in children such as ALL. Stratification in ALL has previously relied on a combination of genetic tests which is time consuming and expensive. In recent studies categorising subtypes of ALL, OGM increased the detection rate and cytogenetic resolution and abrogated the need for cascade testing using multiple cytogenetic techniques, resulting in reduced turnaround times and cost saving (Neveling et al., Reference Neveling, Mantere, Vermeulen, Oorsprong, van Beek, Kater-Baats, Pauper, van der Zande, Smeets, Weghuis, Stevens-Kroef and Hoischen2021; Rack et al., Reference Rack, De Bie, Ameye, Gielen, Demeyer, Cools, De Keersmaecker, Vermeesch, Maertens, Segers, Michaux and Dewaele2022). OGM has also been applied to other areas of paediatrics to improve the diagnostic yield including neurodevelopmental paediatrics (Shieh et al., Reference Shieh, Penon-Portmann, Wong, Levy-Sakin, Verghese, Slavotinek, Gallagher, Mendelsohn, Tenney, Beleford, Perry, Chow, Sharo, Brenner, Qi, Yu, Klein, Martin, Kwok and Boffelli2020), myotonic dystrophy (Otero et al., Reference Otero, Poukalov, Hildebrandt, Thornton, Jinnai, Fujimura, Kimura, Hagerman, Sampson, Day and Wang2021), disorders of sex development (Barseghyan et al., Reference Barseghyan, Délot and Vilain2018) and with the potential for widespread application for prenatal diagnostic testing (Sahajpal et al., Reference Sahajpal, Barseghyan, Kolhe, Hastie and Chaubey2021). Given the extended capabilities of OGM, many undiagnosed conditions in paediatrics will be elucidated using this new technique and other future developments as we enter an era of next-generation cytogenetics (Mantere et al., Reference Mantere, Neveling, Pebrel-Richard, Benoist, van der Zande, Kater-Baats, Baatout, van Beek, Yammine and Oorsprong2020).

Developing biomarkers and targeted therapies to treat childhood diseases

Proteomics

While advances in genomics and cytogenetics are revolutionising our understanding of diseases in childhood, these advances should not be considered in isolation. A systems biology or systems medicine approach is used to consider genetic mutations in the context of or interaction with other biological systems. In vitro and in vivo functional studies examine the translational effect of genetic mutations at a molecular level. Translational studies may include the large-scale impact on protein synthesis and function (proteomics) (Sahajpal et al., Reference Sahajpal, Barseghyan, Kolhe, Hastie and Chaubey2021) and metabolic function within the biological system (metabolomics). It is through the study of proteins within the sphere of proteomics that protein modifications can be characterised, and the targets of drugs identified. The value of proteomics within the context of precision diagnostics in children and adults is the understanding of the post-translational modifications that proteins undergo in response to a variety of intracellular and extracellular signals. Protein phosphorylation is important in protein signalling and disruption of protein phosphorylation due to alteration in protein kinases or phosphatases can lead to oncogenesis (Graves and Haystead, Reference Graves and Haystead2002). The process of cell growth, programmed cell death and the decision to proceed through the cell cycle are all regulated by signal transduction through protein complexes (Hunter, Reference Hunter1995). Dysregulation of these processes results in potential cancer development. Disruption of protein localisation can also have a profound effect on cellular function and can result in diseases in childhood such as cystic fibrosis (Graves and Haystead, Reference Graves and Haystead2002). Detailed knowledge about the structure, function, post-translational modifications, localisation, compartmentalisation and protein–protein interactions within cells supports the development of targeted drug therapies and the use of biomarkers to determine disease development and for monitoring treatment effect (Wilkins et al., Reference Wilkins, Sanchez, Gooley, Appel, Humphery-Smith, Hochstrasser and Williams1996; García-Foncillas et al., Reference García-Foncillas, Argente, Bujanda, Cardona, Casanova, Fernández-Montes, Horcajadas, Iñiguez, Ortiz, Pablos and Pérez Gómez2021). However, proteomics provides an added layer of complexity as protein expression is altered by chronicity and environmental conditions (Al-Amrani et al., Reference Al-Amrani, Al-Jabri, Al-Zaabi, Alshekaili and Al-Khabori2021). To enable drug development and the development of novel biomarkers, public resources of curated signal transduction pathways have been developed (Chatr-aryamontri et al., Reference Chatr-aryamontri, Ceol, Palazzi, Nardelli, Schneider, Castagnoli and Cesareni2007; Mi et al., Reference Mi, Guo, Kejariwal and Thomas2007; Schaefer et al., Reference Schaefer, Anthony, Krupa, Buchoff, Day, Hannay and Buetow2009; Kandasamy et al., Reference Kandasamy, Mohan, Raju, Keerthikumar, Kumar, Venugopal, Telikicherla, Navarro, Mathivanan, Pecquet, Gollapudi, Tattikota, Mohan, Padhukasahasram, Subbannayya, Goel, Jacob, Zhong, Sekhar, Nanjappa, Balakrishnan, Subbaiah, Ramachandra, Rahiman, Prasad, Lin, Houtman, Desiderio, Renauld, Constantinescu, Ohara, Hirano, Kubo, Singh, Khatri, Draghici, Bader, Sander, Leonard and Pandey2010; Croft et al., Reference Croft, O’Kelly, Wu, Haw, Gillespie, Matthews, Caudy, Garapati, Gopinath, Jassal, Jupe, Kalatskaya, Mahajan, May, Ndegwa, Schmidt, Shamovsky, Yung, Birney, Hermjakob, D’Eustachio and Stein2011; Kanehisa et al., Reference Kanehisa, Goto, Sato, Furumichi and Tanabe2012; Kerrien et al., Reference Kerrien, Aranda, Breuza, Bridge, Broackes-Carter, Chen, Duesbury, Dumousseau, Feuermann, Hinz, Jandrasits, Jimenez, Khadake, Mahadevan, Masson, Pedruzzi, Pfeiffenberger, Porras, Raghunath, Roechert, Orchard and Hermjakob2012; Franceschini et al., Reference Franceschini, Szklarczyk, Frankild, Kuhn, Simonovic, Roth, Lin, Minguez, Bork, von Mering and Jensen2013; Holman et al., Reference Holman, Dasari and Tabb2013; Schmidt et al., Reference Schmidt, Forne and Imhof2014). Proteomic-derived biomarker development is classified as diagnostic, predictive and prognostic, based on their uses.

Diagnostic biomarkers indicate if a patient has a specific disease, predictive biomarkers can predict the response to therapy, and prognostic biomarkers support the prediction of the clinical outcomes. Paediatric sepsis provides an example underpinning the value of using a precision diagnostic approach that sub-categorises patients to allow directed therapy to improve outcome and predict prognosis. Sepsis is a leading cause of mortality in children worldwide (Glaab et al., Reference Glaab, Baudot, Krasnogor, Schneider and Valencia2012). Given the heterogeneity of presentation in paediatric septic shock caused by viral or bacterial pathogens, this acts as an excellent model to demonstrate the value of proteomics in relation to precision diagnostic, predictive and prognostic biomarkers in children. Over a decade ago, whole blood RNA was first used to identify genes that were up- and down-regulated in a cohort of paediatric patients with septic shock, demonstrating that these patients had a unique gene expression signature (Wong et al., Reference Wong, Shanley and Sakthivel2007; Liu et al., Reference Liu, Johnson, Cousens, Perin, Scott, Lawn, Rudan, Campbell, Cibulskis, Li, Mathers and Black2012). Early work identified three subclasses of children with septic shock who were defined by age, illness severity and complications classified by 100 genes; younger patients were more likely to have a higher level of illness severity, a higher degree of organ failure and a higher mortality rate (Cvijanovich et al., Reference Cvijanovich, Shanley, Lin, Allen, Thomas, Checchia, Anas, Freishtat, Monaco, Odoms, Sakthivel and Wong2008; Wong et al., Reference Wong, Cvijanovich and Lin2009). Further work in this field has led to the categorisation of children with septic shock into two endotypes which through iterations are now classified by only four genes (Wong et al., Reference Wong, Atkinson, Cvijanovich, Anas, Allen, Thomas, Bigham, Weiss, Fitzgerald, Checchia, Meyer, Quasney, Hall, Gedeit, Freishtat, Nowak, Raj, Gertz and Lindsell2011, Reference Wong, Cvijanovich, Anas, Allen, Thomas, Bigham, Weiss, Fitzgerald, Checchia, Meyer, Shanley, Quasney, Hall, Gedeit, Freishtat, Nowak, Shekhar, Gertz, Dawson, Howard, Harmon, Beckman, Frank and Lindsell2015). Of note, endotype A was noted to have repression of genes corresponding to glucocorticoid receptor signalling and thus adaptive immunity, and use of adjunctive corticosteroids in this group was associated with a 4-fold increase in mortality, thus helping to determine which children with septic shock would benefit from adjunctive corticosteroids and which would not (Wong et al., Reference Wong, Atkinson, Cvijanovich, Anas, Allen, Thomas, Bigham, Weiss, Fitzgerald, Checchia, Meyer, Quasney, Hall, Gedeit, Freishtat, Nowak, Raj, Gertz and Lindsell2011, Reference Wong, Sweeney and Lindsell2017). In addition, multiple serum protein biomarkers with known biological mechanisms in combination with mRNA biomarkers have been used in mortality risk stratification of children with septic shock (Wong et al., Reference Wong, Atkinson and Cvijanovich2016). Combining metabolomic and inflammatory protein mediator profiling early after presentation of paediatric sepsis may differentiate children with sepsis requiring intensive care from those with or without sepsis who can be safely cared for without intensive intervention, thus enabling diagnostic and triage decisions in children with sepsis (Wong et al., Reference Wong, Cvijanovich, Anas, Allen, Thomas, Bigham, Weiss, Fitzgerald, Checchia, Meyer, Quasney, Hall, Gedeit, Freishtat, Nowak, Raj, Gertz, Grunwell and Lindsell2017). Recent work has also helped to define the presence of bacterial or viral pathogens based upon the presence of biomarkers including procalcitonin, neutrophil gelatinase-associated lipocalin-2 and resistin (Mickiewicz et al., Reference Mickiewicz, Thompson, Blackwood, Jenne, Winston, Vogel and Joffe2015) with further advances using host RNA signatures to further discriminate between bacterial and viral infection (Nijman et al., Reference Nijman, Oostenbrink, Moll, Casals-Pascual, von Both, Cunnington, De, Eleftheriou, Emonts, Fink, van der Flier, de Groot, Kaforou, Kohlmaier, Kuijpers, Lim, Maconochie, Paulus, Martinon-Torres, Pokorn, Romaine, Calle, Schlapbach, Smit, Tsolia, Usuf, Wright, Yeung, Zavadska, Zenz, Levin, Herberg and Carrol2021). Other developments in this field include the use of proteomic profiling to distinguish late onset sepsis and necrotising enterocolitis in neonates (Pennisi et al., Reference Pennisi, Moniri, Miscourides, Miglietta, Moser, Habgood-Coote, Herberg, Levin, Kaforou, Rodriguez-Manzano and Georgiou2022), potentially predictive, early diagnostic and prognostic metabolomic and proteomic biomarkers in neonatal necrotising enterocolitis (Chatziioannou et al., Reference Chatziioannou, Wolters, Sarafidis, Thomaidou, Agakidis, Govorukhina, Kuivenhoven, Bischoff and Theodoridis2018), and the combination of metabolomic, transcriptomic, genomic and proteomic profiles as an ‘integrated-omics’ systems medicine approach to the stratification, diagnosis, treatment and prognostics in paediatric sepsis (Agakidou et al., Reference Agakidou, Agakidis, Gika and Sarafidis2020). Outside the field of neonatal and paediatric sepsis, there are other examples where proteomics has the potential to further our understanding of conditions in children. In the field of paediatric allergy, novel proteomics methods have been used to identify the presence of 36 digested cow’s milk proteins in breast milk which would be missed by immunochemical methods, and thus may help to improve our understanding of cow’s milk protein allergy in exclusively breast-fed infants and to support new approaches to allergy prevention (Zhu et al., Reference Zhu, Garrigues, Van den Toorn, Stahl and Heck2019). Recently, proteomics profiling as part of a multi-omics approach has been used to predict neurodegeneration in children with Down’s syndrome pointing to a disruption of IGF1 signalling as a potential contributor to or biomarker to the neurodegenerative process, again potentially providing novel pathways for targeted treatments (Araya et al., Reference Araya, Kinning, Coughlan, Smith, Granrath, Enriquez-Estrada, Worek, Sullivan, Rachubinski, Wolter-Warmerdam, Hickey, Galbraith, Potter and Espinosa2022). In paediatric oncology, proteogenomic studies are furthering our understanding of relapse and treatment resistance in children with acute myeloid leukaemia, providing new approaches to predict relapse during disease progression, novel biomarkers and new targets for novel drug therapies (Stratmann et al., Reference Stratmann, Vesterlund, Umer, Eshtad, Skaftason, Herlin, Sundström, Eriksson, Höglund, Palle, Abrahamsson, Jahnukainen, Munthe-Kaas, Zeller, Tamm, Lindskog, Cavelier, Lehtiö and Holmfeldt2022). Other areas where proteomics provides value to prediction, diagnostics and personalised interventions include chronic kidney disease and peritoneal dialysis in children (Cummins et al., Reference Cummins, Korte, Bhayana, Merchant, Barati, Smoyer and Klein2022; Trincianti et al., Reference Trincianti, Meleca, La Porta, Bruschi, Candiano, Garbarino, Kajana, Preda, Lugani, Ghiggeri, Angeletti, Esposito and Verrina2022), drug metabolism and personalised dose optimisation in children (Streekstra et al., Reference Streekstra, Russel, van de Steeg and de Wildt2021; van Groen et al., Reference van Groen, Allegaert, Tibboel and de Wildt2022) and the use of multi-omics derived biomarkers to develop prediction models in children with asthma to guide therapy (Golebski et al., Reference Golebski, Kabesch, Melén, Potočnik, van Drunen, Reinarts, Maitland-van der Zee and Vijverberg2020; Kang et al., Reference Kang, Ahn, Lee, Yeom, Kim and Hong2022). Proteomics either in isolation or part of a multi-omics approach has revolutionised our ability to develop individual disease profiling across multiple areas of paediatrics with new diagnostic, predictive and prognostic methodologies, and new targets for novel drug development.

Precision diagnostics for targeted drug therapies

An understanding of genetic causes of specific diseases has resulted in the recent increase in the development of gene therapies in children regulated under the guidelines for advanced therapy medicinal products. Broadly, gene therapies were previously categorised into ex vivo and in vivo (Langley and Wong, Reference Langley and Wong2017). Ex vivo is the route taken predominantly for the gene modification of bone marrow-derived cells and epidermal sheets. Ex vivo gene therapies are based upon the process of extracting immature bone marrow cells from the patient, employing a viral vector to integrate a functional copy of the gene into the genome of the target cells. The genetically modified cells are then re-administered to the patient in the form of an autologous gene-modified cell transplant. Examples of ex vivo gene therapies include the treatment of primary immune deficiencies and metabolic disorders (Qasim et al., Reference Qasim, Gaspar and Thrasher2007; Buckland and Bobby Gaspar, Reference Buckland and Bobby Gaspar2014) with more recent approvals granted for rare diseases such as metachromatic leukodystrophy (treated with atidarsagene autotemcel) (Rivat et al., Reference Rivat, Santilli, Gaspar and Thrasher2012) and cerebral adrenoleukodystrophy (treated with elivaldogene autotemcel) (Fumagalli et al., Reference Fumagalli, Calbi, Natali Sora, Sessa, Baldoli, Rancoita, Ciotti, Sarzana, Fraschini, Zambon, Acquati, Redaelli, Attanasio, Miglietta, De Mattia, Barzaghi, Ferrua, Migliavacca, Tucci, Gallo, Del Carro, Canale, Spiga, Lorioli, Recupero, Fratini, Morena, Silvani, Calvi, Facchini, Locatelli, Corti, Zancan, Antonioli, Farinelli, Gabaldo, Garcia-Segovia, Schwab, Downey, Filippi, Cicalese, Martino, Di Serio, Ciceri, Bernardo, Naldini, Biffi and Aiuti2022). The first UK patient to receive gene therapy in the UK celebrated his 21st birthday last year after having been treated for severe combined immunodeficiency (Keam, Reference Keam2021). In vivo gene therapy primarily uses a viral vector (commonly the adeno-associated virus) to deliver the therapy directly to the patient rather than the transplant of gene-corrected cells. Glybera (alipogene tiparvovec) was the first gene therapy treatment to receive an European Commission marketing authorisation and the first gene therapy to be approved anywhere in the world, was used to treat lipoprotein lipase deficiency, an inherited condition with an incidence of 1/500,000. This therapy has since been withdrawn due to the rarity of the disease lack of cost-effectiveness (Great Ormond Street Hospital for Children (GOSH), 2021; Kastelein et al., Reference Kastelein, Ross and Hayden2013). There are currently five gene therapy treatments approved in Europe – Luxturna (for individuals with an inherited retinal disease caused by mutations in both copies of the RPE65 gene), Zolgensma (to treat spinal muscular atrophy), two chimeric antigen receptor T cell therapies (Yescarta – to treat non-Hodgkin lymphoma and Kymriah for the treatment of adult patients with relapsed or refractory follicular lymphoma) and Strimvelis (the gamma-retrovirus for adenosine deaminase-severe combined immunodeficiency). Multiple other clinical trials are ongoing to identify candidate genes amenable to in vivo gene therapies (Ylä-Herttuala, Reference Ylä-Herttuala2012; Mendell et al., Reference Mendell, Al-Zaidy and Rodino-Klapac2021). The recent advent of genome editing uses a different approach to correct genetic differences by introducing molecular tools to change existing DNA. Approaches include the use of zinc finger nucleases (Lee et al., Reference Lee, Cho, Huang, Xu, Quach, Hu and Wong2021), transcription activator-like effector nucleases (Carroll, Reference Carroll2011) and clustered regularly interspaced short palindromic repeats (Liu et al., Reference Liu, Gaj, Yang, Wang, Shui, Kim, Kanchiswamy, Kim and Barbas2015), which has catalysed the implementation of new clinical trials of gene editing to cure rare and common diseases in children that otherwise result in death in childhood or early adulthood (Lee et al., Reference Lee, Jeong, Lee, Jung, Shin, Kim, Lee, Jung, Kim, Kim and Kim2018, Reference Lee, Gillam, Visvanathan, Hansford and McCarthy2021).

Biomarker-driven directed therapies in the treatment of cancer in children are still early in development. A recent systematic review reporting on the clinical utility of precision medicine in the treatment of paediatric cancer has reported on the use of molecular techniques such as array comparative hybridisation, immunohistochemistry, next-generation sequencing, RNA sequencing, single nucleotide polymorphism array, targeted panel-based sequencing, whole-exome sequencing and WGS to identify genomic targets, which have guided the allocation of targeted drugs (Salsman and Dellaire, Reference Salsman and Dellaire2017). Genes which are transcribed in any one condition are known as the transcriptome; the process of determining the genetic codes contained in the transcriptome and their relative proportions is known as transcriptome sequencing or transcriptomics. Of note, the value of understanding the transcriptome has been highlighted through the increased benefit of identifying additional therapeutic targets that were not identified by genomic analysis alone (Marks et al., Reference Marks, Oberg, Pendrick, Sireci, Glasser, Coval, Zylber, Chung, Pang, Turk, Hsiao, Mansukhani, Glade Bender, Kung and Sulis2017; Uddin et al., Reference Uddin, Rudin and Sen2020). Targetable mutations were found in 48.0% of patients, with 41.7% who received targeted drugs demonstrating an objective response. However, accessibility and thus inequity of access to targeted oncological therapies were cited as problematic, with only 27% of patients receiving targeted treatments, highlighting the gulf between access to precision diagnostics and subsequent therapies (Salsman and Dellaire, Reference Salsman and Dellaire2017). Challenges cited included the need to access drugs off-licence due to lack of paediatric dosing schedules, lack of access to clinical trials at their treating centre or ineligibility owing to advanced disease or trial restriction to adult patients or the dependency on compassionate access facilitated by pharmaceutical companies (Salsman and Dellaire, Reference Salsman and Dellaire2017). Moreover, heterogeneity in outcome reporting across clinical trials may lead to bias in the interpretation of treatment effect. This underpins the need for consistency in clinical trial outcomes to clearly define the clinical and economic value of a precision diagnostics approach to targeted therapy. Large molecular databases and global collaborative clinical trials have been assembled to help demonstrate the value of targeted therapies which will in turn enhance our understanding in the future and improve access to novel therapies (Pincez et al., Reference Pincez, Clément, Lapouble, Pierron, Kamal, Bieche, Bernard, Fréneaux, Michon, Orbach, Aerts, Pacquement, Bourdeaut, Jiménez, Thébaud, Oudot, Vérité, Taque, Owens, Doz, Le Tourneau, Delattre and Schleiermacher2000; Brien et al., Reference Brien, Valerio and Armstrong2016; Harttrampf et al., Reference Harttrampf, Lacroix, Deloger, Deschamps, Puget, Auger, Vielh, Varlet, Balogh, Abbou, Allorant, Valteau-Couanet, Sarnacki, Gamiche-Rolland, Meurice, Minard-Colin, Grill, Brugieres, Dufour, Gaspar, Michiels, Vassal, Soria and Geoerger2017; Linzey et al., Reference Linzey, Marini, Pasternak, Smith, Miklja, Zhao, Kumar-Sinha, Paul, Harris, Robertson, Hoffman, Chinnaiyan, Mody and Koschmann2018; George et al., Reference George, Izquierdo, Campbell, Koutroumanidou, Proszek, Jamal, Hughes, Yuan, Marshall, Carceller, Chisholm, Vaidya, Mandeville, Angelini, Wasti, Bexelius, Thway, Gatz, Clarke, Al-Lazikani, Barone, Anderson, Tweddle, Gonzalez, Walker, Barton, Depani, Eze, Ahmed, Moreno, Pearson, Shipley, Jones, Hargrave, Jacques, Hubank and Chesler2019; Hansford, Reference Hansford2019; Gojo et al., Reference Gojo, Pavelka, Zapletalova, Schmook, Mayr, Madlener, Kyr, Vejmelkova, Smrcka, Czech, Dorfer, Skotakova, Azizi, Chocholous, Reisinger, Lastovicka, Valik, Haberler, Peyrl, Noskova, Pál, Jezova, Veselska, Kozakova, Slaby, Slavc and Sterba2020).

A digital approach to precision diagnostics and medicine in paediatrics

The value of population data

The focus on precision diagnostics in paediatrics has largely focused on the value of data derived from ‘omics’ profiling as a means of supporting diagnoses, risk stratification, directing therapy (pharmacogenomics and pharmacometabolomics) and determining prognostic indicators. While this approach is revolutionising the way patients are managed, a systems medicine approach should also consider data relating to patient populations and external factors that may impact on patient diagnosis, monitoring and care. While precision in the initial diagnosis enhances a personalised approach to therapy, longitudinal patient monitoring facilitates a dynamic precision approach to ‘diagnosing’ new or recurrent issues. As medical device development in paediatrics and child health gathers momentum (Vo et al., Reference Vo, Parsons and Seibel2020), the value of personalised and population data to direct patient management is attracting attention. For example, large data sets relating to growth in children combined with automated systems for monitoring linear growth are facilitating the early identification of growth disorders of children at a greater frequency while reducing the number of referrals of children with normal growth parameters (Dimitri, Reference Dimitri2019). For patients diagnosed with disorders of growth requiring growth hormone therapy, large-scale data acquisition using a growth hormone delivery device with a connected monitoring platform (easypod™), which automatically transmits adherence data via an online portal (easypod™ connect), can be used to predict an individual’s response to therapy relative to a population undergoing the same treatment, by integrating methodologies such as machine learning. This facilitates clinical decision support to modify therapy in individual patients (Sankilampi et al., Reference Sankilampi, Saari, Laine, Miettinen and Dunkel2013). This approach of using patient population data to ‘diagnose’ poor adherence to therapy has been a catalyst to develop a digital ecosystem to support service users and service providers in the management of growth disorders requiring growth hormone therapy. This includes the development of online educational materials to support digital literacy and patient management (Dimitri et al., Reference Dimitri, Fernandez-Luque, Koledova, Bagha and Shabbir2000; Su et al., Reference Su, Malik, Jheeta, Lin, Su, Koledova and Graham2022), digital monitoring of patient therapy, adherence and preferences (Boman et al., Reference Boman, Fernandez-Luque, Koledova, Kause and Lapatto2021; Spataru et al., Reference Spataru, Quarteroni, Arnaud, van Dommelen, Koledova and Le Masne2021; Spataru et al., Reference Spataru, van Dommelen, Arnaud, Le Masne, Quarteroni and Koledova2022; van Dommelen et al., Reference van Dommelen, Koledova and Wit2018; Koledova et al., Reference Koledova, Tornincasa and van Dommelen2020) and the development of a framework to guide future digital developments including automated referral pathways, enhanced digital communication, digital medical and psychological support, gamification to support adherence to therapy, access to digital resources, digital reporting of patient reported outcomes and safety and assessment reporting (Dimitri et al., Reference Dimitri, Fernandez-Luque, Banerjee, Bergadá, Calliari, Dahlgren, de Arriba, Lapatto, Reinehr, Senniappan, Thomas-Teinturier, Tsai, Anuar Zaini, Bagha and Koledova2021).

The application of machine learning in paediatric diagnosis

The advent of machine learning has facilitated the use of large patient data sets or ‘training sets’ to generate and refine predictive models. Large data sets already exist in multiple areas of medicine, and these are now being employed to predict disease in children. Machine learning has superior capabilities to traditional data analytics methods based upon the ability to rapidly process large volumes of complex data, to explore and extrapolate data relationships through pattern recognition that are not recognisable through other methods and improving efficiency and accuracy through data acquisition. Deep learning lets the data train the computer leading to predictive models that become stronger as more data are added. Imaging data sets that are being used in paediatrics to predict abnormalities have been frequently reported in the literature in the last few years with examples including (but not limited to) the detection of abnormalities in chest radiographs (Chen et al., Reference Chen, Yu, Chen, Lin, Lee, Chen, Jiang, Su, Tsai, Tsai, Tsai and Lu2020; Padash et al., Reference Padash, Mohebbian, Adams, Henderson and Babyn2022), the diagnosis of effusions in elbow joints (Huhtanen et al., Reference Huhtanen, Nyman, Doncenco, Hamedian, Kawalya, Salminen, Sequeiros, Koskinen, Pudas, Kajander, Niemi, Hirvonen, Aronen and Jafaritadi2022), the detection of intracranial pathology on CT imaging, defining abnormalities as critical or non-critical (Titano et al., Reference Titano, Badgeley, Schefflein, Pain, Su, Cai, Swinburne, Zech, Kim, Bederson, Mocco, Drayer, Lehar, Cho, Costa and Oermann2018), the assessment of left ventricular function from birth to 18 years and the diagnosis of coronary artery lesions in Kawasaki disease using echocardiography (Lee et al., Reference Lee, Eun, Hwang and Eun2022; Zuercher et al., Reference Zuercher, Ufkes, Erdman, Slorach, Mertens and Taylor2022) and the assessment of paediatric brain tumours (Grist et al., Reference Grist, Withey, Bennett, Rose, MacPherson, Oates, Powell, Novak, Abernethy, Pizer, Bailey, Clifford, Mitra, Arvanitis, Auer, Avula, Grundy and Peet2021; Huang et al., Reference Huang, Shlobin, Lam and DeCuypere2022). However, challenges remain in achieving an acceptable diagnostic accuracy and the acquisition of adequate training data sets in cohorts, particularly in relation to rare diseases due to the size of the patient populations. Machine learning has been applied to continuous conventional electroencephalography to determine the diagnostic accuracy of detecting and monitoring seizures in neonates. The artificial intelligence (AI) platform did not improve the number of neonates diagnosed with seizures, although the quantification of seizure burden was greater, thus providing a more accurate means of monitoring seizure frequency and duration (Pavel et al., Reference Pavel, Rennie, de Vries, Blennow, Foran, Shah, Pressler, Kapellou, Dempsey, Mathieson, Pavlidis, van Huffelen, Livingstone, Toet, Weeke, Finder, Mitra, Murray, Marnane and Boylan2020). The DeepGestalt platform utilising computer vision and deep-learning algorithms for facial image analysis has shown great potential in the phenotypic evaluation of syndromes including the initial syndromic diagnosis and the ability to subclassify different genetic subtypes within the same syndromic diagnosis (Gurovich et al., Reference Gurovich, Hanani, Bar, Nadav, Fleischer, Gelbman, Basel-Salmon, Krawitz, Kamphausen, Zenker, Bird and Gripp2019). Patient population data have also been used to detect the earlier presentation of more common presentations in paediatrics such as neonatal sepsis (Masino et al., Reference Masino, Harris, Forsyth, Ostapenko, Srinivasan, Bonafide, Balamuth, Schmatz and Grundmeier2019), the risk stratification of infants with bronchiolitis to promptly identify infants at risk of deterioration (Raita et al., Reference Raita, Camargo, Macias, Mansbach, Piedra, Porter, Teach and Hasegawa2020), improving the prediction of clinical outcomes in children presenting to the emergency department with the benefit of better identifying critically ill children while reducing the over triaging of children who are less ill (Raita et al., Reference Raita, Camargo, Macias, Mansbach, Piedra, Porter, Teach and Hasegawa2020) and predicting the need for hospitalisation of children with asthma (Patel et al., Reference Patel, Chamberlain and Chamberlain2018). Notably, this last example also demonstrates the value of using environmental data including weather data, population influenza patterns and socioeconomic status to improve predictability. The value of data acquisition from social media platforms on smartphones and tables to support the diagnosis of mental health issues in young people and adults such as depression (Reece and Danforth, Reference Reece and Danforth2017), post-traumatic stress disorder (Reece et al., Reference Reece, Reagan, Lix, Dodds, Danforth and Langer2017) and schizophrenia (Hänsel et al., Reference Hänsel, Lin, Sobolev, Muscat, Yum-Chan, De Choudhury, Kane and Birnbaum2021) highlights the value of ‘non-health’ data to support diagnoses. Given that children and young people are high users of technology and social media, it is important to consider the value of personalised data on commercially available platforms in the future to inform on health, well-being and disease. For example, Instagram posts have been used as a predictor of physical activity in adults aged 18–30 years (Liu et al., Reference Liu, Perdew, Lithopoulos and Rhodes2021). A comprehensive review of machine learning in paediatrics is covered elsewhere (Clarke et al., Reference Clarke, Parmesar, Saleem and Ramanan2022), supporting the value of AI in the field of precision diagnostics in paediatrics. As electronic patient records become the global norm, it is likely that the application AI will become fundamental to improve diagnosis and clinical outcomes. However, large and accurate data sets will be required to ensure accurate outputs from machine learning models, and legal and governance structures will need to be in place to regulate the use of these data. Given the relative paucity of large paediatric data sets, interoperability across systems and continental or global data sharing of high-quality data may be required to optimise outputs but will also need to be viewed in the context of generalisability across different healthcare systems.

Future perspectives

Advances in science, technology and data acquisition and analytics have already revolutionised paediatric healthcare. Adopting the ‘4P’ systems medicine approach (Hood et al., Reference Hood, Heath, Phelps and Lin2004; Flores et al., Reference Flores, Glusman, Brogaard, Price and Hood2013) to ensure healthcare is preventive, predictive, participatory and personalised will rely on the combination of multiple data sources and methodologies to maintain health and prevent and predict disease. The value of combining ‘omics’ data has advanced the science of molecular precision medicine to understand the molecular basis of disease, the risk of developing it and its evolution and pathogenesis in children; the speed of technological advances in genomics, proteomics, transcriptomics and AI will further improve this (Williams et al., Reference Williams, Liu, Regner, Jotterand, Liu and Liang2018; García-Foncillas et al., Reference García-Foncillas, Argente, Bujanda, Cardona, Casanova, Fernández-Montes, Horcajadas, Iñiguez, Ortiz, Pablos and Pérez Gómez2021).

The future of precision diagnostics in paediatrics will in part rely on data acquisition that has not been commonly used in medicine before from commercial and social media platforms, and from environmental sources, knowing that the lifestyle, demographic and environmental factors play a key role in the presentation and course of disease. Personalised platforms on mobile phone and tablets, digital monitoring systems within homes and remote sensing technologies can gather data on personal habits, lifestyle, physical activity and diet, and environmental data such as climate data can be derived from institutional sources. Collectively, these factors are encapsulated in the concept of the ‘exposome’. The ‘exposome’ has been proposed as a new paradigm to encompass the totality of human environmental (non-genetic) exposures from conception onwards (Wild, Reference Wild2012; Vrijheid, Reference Vrijheid2014). Thus, the exposome will play a fundamental part in precision diagnostics through the life course. In this context, there is a value in collecting data from early life to truly reflect response to changes in the exposome which in turn support disease prevention, predicting the future risk of developing a disease and supporting the management of disease in an environmental context once established. To facilitate this, advances in sensing devices and other technologies will facilitate collection and remote access to these data (Martin Sanchez et al., Reference Martin Sanchez, Gray, Bellazzi and Lopez-Campos2014). In addition to the challenges of collecting and extrapolating data, data analytics and combining data sets, the context in which these data are derived must be considered. Despite the inherent affinity children and young people have with technology, there is a reluctance in this population to simply ‘hand over’ data, and data acquisition must be contextualised to social context. Children and young people are reluctant to use technology to collect health data if this does not fit in with social norms, thus risking discrimination. Concerns have been raised surrounding data sharing, use and confidentiality of personal information, with children and young people needing reassurance that their data are being used by trusted organisations, that it is stored safely and securely, with a desire to control their data and privacy, and to minimise the risk of misinterpretation of their health data, particularly in the context of them having a health-related problem (Blower et al., Reference Blower, Swallow, Maturana, Stones, Phillips, Dimitri, Marshman, Knapp, Dean, Higgins, Kellar, Curtis, Mills and Martin-Kerry2020). Participation is framed as a central tenet of personalised medicine. As we move in the direction of whole-scale personalised medicine that involves the collection and storage of data in children, particularly in relation to the future predictability or certainty of disease, the ethical and legal considerations around informed consent, data protection, autonomy and privacy must be considered carefully, particularly for those who are not old enough to express their views. There are potential legal and ethical ramifications from extensive testing and use of data that may have unintended consequences, for example, the identification of a secondary disease that does not manifest until adulthood, that in turn may limit testing in children. Thus, guidelines on how to pursue children’s participation in personalised medicine would be of benefit in the future (Ó Cathaoir, Reference Ó Cathaoir2021).

Conclusion

The way data is changing health and healthcare in the 21st century could be likened to the impact that the introduction of antibiotics had on bacterial disease in the early 20th century – it will save and improve many lives. We have already seen significant advances in diagnosis, drug discovery and interventions in healthcare as a result of advances in the ‘omics’ and cytogenetics. The future acquisition and combination of data from multiple sources including data from individuals, their lifestyle behaviours and their immediate and wider environments will add to our understanding of the manifestation of health and disease over time. A life course approach starting from birth or even in the prenatal period is required to understand how human development and changes in the environment lead to future health and ill-health, and how environmental and physical factors impact on congenital and acquired diseases of childhood. As we advance in the field of personalised medicine, we need to consider a paradigm shift in precision diagnostics to incorporate the need to ‘sustain health’ in addition to accurately diagnosing disease. In this respect, this shift in diagnostics should encompass the need to ‘diagnose’ good health. The principle of ‘start early and change more’ underpins the value of applying personalised medicine early in life for a better future.

Open peer review

To view the open peer review materials for this article, please visit http://doi.org/10.1017/pcm.2023.4.

Competing interest

The author declares no conflict of interest.

References

Agakidou, E, Agakidis, C, Gika, H and Sarafidis, K (2020) Emerging biomarkers for prediction and early diagnosis of necrotizing enterocolitis in the era of metabolomics and proteomics. Frontiers in Pediatrics 8, 602255.CrossRefGoogle ScholarPubMed
Al-Amrani, S, Al-Jabri, Z, Al-Zaabi, A, Alshekaili, J and Al-Khabori, M (2021) Proteomics: Concepts and applications in human medicine. World Journal of Biological Chemistry 12(5), 5769.Google Scholar
Araya, P, Kinning, KT, Coughlan, C, Smith, KP, Granrath, RE, Enriquez-Estrada, BA, Worek, K, Sullivan, KD, Rachubinski, AL, Wolter-Warmerdam, K, Hickey, F, Galbraith, MD, Potter, H and Espinosa, JM (2022) IGF1 deficiency integrates stunted growth and neurodegeneration in down syndrome. Cell Reports 41(13), 111883.CrossRefGoogle ScholarPubMed
Barseghyan, H, Délot, EC and Vilain, E (2018) New technologies to uncover the molecular basis of disorders of sex development. Molecular and Cellular Endocrinology 468, 6069.CrossRefGoogle ScholarPubMed
Blower, S, Swallow, V, Maturana, C, Stones, S, Phillips, R, Dimitri, P, Marshman, Z, Knapp, P, Dean, A, Higgins, S, Kellar, I, Curtis, P, Mills, N and Martin-Kerry, J (2020) Children and young people’s concerns and needs relating to their use of health technology to self-manage long-term conditions: A scoping review. Archives of Disease in Childhood 105(11), 10931104.Google Scholar
Boman, N, Fernandez-Luque, L, Koledova, E, Kause, M and Lapatto, R (2021) Connected health for growth hormone treatment research and clinical practice: Learnings from different sources of real-world evidence (RWE)-large electronically collected datasets, surveillance studies and individual patients’ cases. BMC Medical Informatics and Decision Making 21(1), 136.CrossRefGoogle ScholarPubMed
Brien, GL, Valerio, DG and Armstrong, SA (2016) Clinical insights gained by refining the 2016 WHO classification of diffuse gliomas with: EGFR amplification, TERT mutations, PTEN deletion and MGMT methylation. Cancer Cell 29, 464476.Google Scholar
Buckland, KF and Bobby Gaspar, H (2014) Gene and cell therapy for children–New medicines, new challenges? Advanced Drug Delivery Reviews 73(100), 162169.Google ScholarPubMed
Carroll, D (2011) Genome engineering with zinc-finger nucleases. Genetics 188(4), 773782.CrossRefGoogle ScholarPubMed
Chatr-aryamontri, A, Ceol, A, Palazzi, LM, Nardelli, G, Schneider, MV, Castagnoli, L and Cesareni, G (2007) MINT: The molecular INTeraction database. Nucleic Acids Research 35, D572D574.CrossRefGoogle ScholarPubMed
Chatziioannou, AC, Wolters, JC, Sarafidis, K, Thomaidou, A, Agakidis, C, Govorukhina, N, Kuivenhoven, JA, Bischoff, R and Theodoridis, G (2018) Targeted LC-MS/MS for the evaluation of proteomics biomarkers in the blood of neonates with necrotizing enterocolitis and late-onset sepsis. Analytical and Bioanalytical Chemistry 410(27), 71637175.Google ScholarPubMed
Chen, KC, Yu, HR, Chen, WS, Lin, WC, Lee, YC, Chen, HH, Jiang, JH, Su, TY, Tsai, CK, Tsai, TA, Tsai, CM and Lu, HH (2020) Diagnosis of common pulmonary diseases in children by X-ray images and deep learning. Scientific Reports 10(1), 17374.Google ScholarPubMed
Clarke, SL, Parmesar, K, Saleem, MA and Ramanan, AV (2022) Future of machine learning in paediatrics. Archives of Disease in Childhood 107(3), 223228.Google ScholarPubMed
Costain, G, Cohn, RD and Malkin, D (2020) Precision child health: An emerging paradigm for Paediatric quality and safety. Current Treatment Options in Pediatrics 6(4), 317324.CrossRefGoogle Scholar
Croft, D, O’Kelly, G, Wu, G, Haw, R, Gillespie, M, Matthews, L, Caudy, M, Garapati, P, Gopinath, G, Jassal, B, Jupe, S, Kalatskaya, I, Mahajan, S, May, B, Ndegwa, N, Schmidt, E, Shamovsky, V, Yung, C, Birney, E, Hermjakob, H, D’Eustachio, P and Stein, L (2011) Reactome: A database of reactions, pathways and biological processes. Nucleic Acids Research Pla 39, D691D697.Google ScholarPubMed
Cummins, TD, Korte, EA, Bhayana, S, Merchant, ML, Barati, MT, Smoyer, WE and Klein, JB (2022) Advances in proteomic profiling of pediatric kidney diseases. Pediatric Nephrology 37(10), 22552265.Google ScholarPubMed
Cvijanovich, N, Shanley, TP, Lin, R, Allen, GL, Thomas, NJ, Checchia, P, Anas, N, Freishtat, RJ, Monaco, M, Odoms, K, Sakthivel, B, Wong, HR; Genomics of Pediatric SIRS/Septic Shock Investigators. (2008) Validating the genomic signature of pediatric septic shock. Physiological Genomics 34(1), 127134.Google ScholarPubMed
Dahlgren, J and Noordam, C (2022) Growth, endocrine features, and growth hormone treatment in Noonan syndrome. Journal of Clinical Medicine 11(7), 2034.CrossRefGoogle ScholarPubMed
Dimitri, P (2019) Child health technology: Shaping the future of paediatrics and child health and improving NHS productivity. Archives of Disease in Childhood 104(2), 184188.CrossRefGoogle ScholarPubMed
Dimitri, P, Fernandez-Luque, L, Banerjee, I, Bergadá, I, Calliari, LE, Dahlgren, J, de Arriba, A, Lapatto, R, Reinehr, T, Senniappan, S, Thomas-Teinturier, C, Tsai, MC, Anuar Zaini, A, Bagha, M and Koledova, E (2021) An eHealth framework for managing pediatric growth disorders and growth hormone therapy. Journal of Medical Internet Research 23(5), e27446.CrossRefGoogle ScholarPubMed
Dimitri, P, Fernandez-Luque, L, Koledova, E, Bagha, M and Shabbir, SA (2021) Massive open online learning – Accelerating knowledge in digital health in the management of children with growth disorders. Hormone Research in Paediatrics 82(Suppl. 1, 94, P2P246.Google Scholar
Flores, M, Glusman, G, Brogaard, K, Price, ND and Hood, L (2013) P4 medicine: How systems medicine will transform the healthcare sector and society. Personalized Medicine 10(6), 565576.Google ScholarPubMed
Franceschini, A, Szklarczyk, D, Frankild, S, Kuhn, M, Simonovic, M, Roth, A, Lin, J, Minguez, P, Bork, P, von Mering, C and Jensen, LJ (2013) STRING v9.1: Protein-protein interaction networks, with increased coverage and integration. Nucleic Acids Research 41, D808D815.CrossRefGoogle ScholarPubMed
Fumagalli, F, Calbi, V, Natali Sora, MG, Sessa, M, Baldoli, C, Rancoita, PMV, Ciotti, F, Sarzana, M, Fraschini, M, Zambon, AA, Acquati, S, Redaelli, D, Attanasio, V, Miglietta, S, De Mattia, F, Barzaghi, F, Ferrua, F, Migliavacca, M, Tucci, F, Gallo, V, Del Carro, U, Canale, S, Spiga, I, Lorioli, L, Recupero, S, Fratini, ES, Morena, F, Silvani, P, Calvi, MR, Facchini, M, Locatelli, S, Corti, A, Zancan, S, Antonioli, G, Farinelli, G, Gabaldo, M, Garcia-Segovia, J, Schwab, LC, Downey, GF, Filippi, M, Cicalese, MP, Martino, S, Di Serio, C, Ciceri, F, Bernardo, ME, Naldini, L, Biffi, A and Aiuti, A (2022) Lentiviral haematopoietic stem-cell gene therapy for early-onset metachromatic leukodystrophy: Long-term results from a non-randomised, open-label, phase 1/2 trial and expanded access. Lancet 399(10322), 372383.CrossRefGoogle ScholarPubMed
García-Foncillas, J, Argente, J, Bujanda, L, Cardona, V, Casanova, B, Fernández-Montes, A, Horcajadas, JA, Iñiguez, A, Ortiz, A, Pablos, JL and Pérez Gómez, MV (2021) Milestones of precision medicine: An innovative, multidisciplinary overview. Molecular Diagnosis & Therapy 25(5), 563576.Google ScholarPubMed
George, SL, Izquierdo, E, Campbell, J, Koutroumanidou, E, Proszek, P, Jamal, S, Hughes, D, Yuan, L, Marshall, LV, Carceller, F, Chisholm, JC, Vaidya, S, Mandeville, H, Angelini, P, Wasti, A, Bexelius, T, Thway, K, Gatz, SA, Clarke, M, Al-Lazikani, B, Barone, G, Anderson, J, Tweddle, DA, Gonzalez, D, Walker, BA, Barton, J, Depani, S, Eze, J, Ahmed, SW, Moreno, L, Pearson, A, Shipley, J, Jones, C, Hargrave, D, Jacques, TS, Hubank, M and Chesler, L (2019) A tailored molecular profiling programme for children with cancer to identify clinically actionable genetic alterations. European Journal of Cancer 121, 224235.CrossRefGoogle ScholarPubMed
Glaab, E, Baudot, A, Krasnogor, N, Schneider, R and Valencia, A (2012) EnrichNet: Network-based gene set enrichment analysis. Bioinformatics 28, i451i457.Google ScholarPubMed
Gojo, J, Pavelka, Z, Zapletalova, D, Schmook, MT, Mayr, L, Madlener, S, Kyr, M, Vejmelkova, K, Smrcka, M, Czech, T, Dorfer, C, Skotakova, J, Azizi, AA, Chocholous, M, Reisinger, D, Lastovicka, D, Valik, D, Haberler, C, Peyrl, A, Noskova, H, Pál, K, Jezova, M, Veselska, R, Kozakova, S, Slaby, O, Slavc, I and Sterba, J (2020) Personalized treatment of H3K27M-mutant pediatric diffuse gliomas provides improved therapeutic opportunities. Frontiers in Oncology 9, 1436.CrossRefGoogle ScholarPubMed
Golebski, K, Kabesch, M, Melén, E, Potočnik, U, van Drunen, CM, Reinarts, S, Maitland-van der Zee, AH, Vijverberg, SJH, and PERMEABLE consortium (2020) Childhood asthma in the new omics era: Challenges and perspectives. Current Opinion in Allergy and Clinical Immunology 20(2), 155161,CrossRefGoogle ScholarPubMed
Graves, PR and Haystead, TA (2002) Molecular biologist’s guide to proteomics. Microbiology and Molecular Biology Reviews 66(1), 3963.CrossRefGoogle ScholarPubMed
Great Ormond Street Hospital for Children (GOSH) (2021). Available at https://www.gosh.org/news/uks-first-gene-therapy-baby-celebrates-21st-birthday/.Google Scholar
Gripp, KW, Schill, L, Schoyer, L, Stronach, B, Bennett, AM, Blaser, S, Brown, A, Burdine, R, Burkitt-Wright, E, Castel, P, Darilek, S, Dias, A, Dyer, T, Ellis, M, Erickson, G, Gelb, BD, Green, T, Gross, A, Ho, A, Holder, JL Jr, Inoue, SI, Jelin, AC, Kennedy, A, Klein, R, Kontaridis, MI, Magoulas, P, McConnell, DB, McCormick, F, Neel, BG, Prada, CE, Rauen, KA, Roberts, A, Rodriguez-Viciana, P, Rosen, N, Rumbaugh, G, Sablina, A, Solman, M, Tartaglia, M, Thomas, A, Timmer, WC, Venkatachalam, K, Walsh, KS, Wolters, PL, Yi, JS, Zenker, M and Ratner, N (2020) The sixth international RASopathies symposium: Precision medicine-from promise to practice. American Journal of Medical Genetics. Part A 182(3), 597606.CrossRefGoogle ScholarPubMed
Grist, JT, Withey, S, Bennett, C, Rose, HEL, MacPherson, L, Oates, A, Powell, S, Novak, J, Abernethy, L, Pizer, B, Bailey, S, Clifford, SC, Mitra, D, Arvanitis, TN, Auer, DP, Avula, S, Grundy, R and Peet, AC (2021) Combining multi-site magnetic resonance imaging with machine learning predicts survival in pediatric brain tumors. Scientific Reports 11(1), 18897.CrossRefGoogle ScholarPubMed
Gurovich, Y, Hanani, Y, Bar, O, Nadav, G, Fleischer, N, Gelbman, D, Basel-Salmon, L, Krawitz, PM, Kamphausen, SB, Zenker, M, Bird, LM and Gripp, KW (2019) Identifying facial phenotypes of genetic disorders using deep learning. Nature Medicine 25(1), 6064.CrossRefGoogle ScholarPubMed
Hänsel, K, Lin, IW, Sobolev, M, Muscat, W, Yum-Chan, S, De Choudhury, M, Kane, JM and Birnbaum, ML (2021) Utilizing Instagram data to identify usage patterns associated with schizophrenia Spectrum disorders. Frontiers in Psychiatry 12, 691327.CrossRefGoogle ScholarPubMed
Hansford, JR (2019) Personalised medicine in paediatric oncology: Ethical practice outside the clinical trial framework? Journal of Paediatrics and Child Health 55(1), 1012.Google ScholarPubMed
Harttrampf, AC, Lacroix, L, Deloger, M, Deschamps, F, Puget, S, Auger, N, Vielh, P, Varlet, P, Balogh, Z, Abbou, S, Allorant, A, Valteau-Couanet, D, Sarnacki, S, Gamiche-Rolland, L, Meurice, G, Minard-Colin, V, Grill, J, Brugieres, L, Dufour, C, Gaspar, N, Michiels, S, Vassal, G, Soria, JC and Geoerger, B (2017) Molecular screening for cancer treatment optimization (MOSCATO-01) in pediatric patients: A single-institutional prospective molecular stratification trial. Clinical Cancer Research 23, 61016112.CrossRefGoogle ScholarPubMed
Holman, JD, Dasari, S and Tabb, DL (2013) Informatics of protein and posttranslational modification detection via shotgun proteomics. Methods in Molecular Biology 1002, 167179.CrossRefGoogle ScholarPubMed
Hood, L, Balling, R and Auffray, C (2012) Revolutionizing medicine in the 21st century through systems approaches. Biotechnology Journal 7(8), 9921001.Google ScholarPubMed
Hood, L, Heath, JR, Phelps, ME and Lin, B (2004) Systems biology and new technologies enable predictive and preventative medicine. Science 306(5696), 640643.CrossRefGoogle ScholarPubMed
Huang, J, Shlobin, NA, Lam, SK and DeCuypere, M (2022) Artificial intelligence applications in pediatric brain tumor imaging: A systematic review. World Neurosurgery 157, 99105.Google ScholarPubMed
Huhtanen, JT, Nyman, M, Doncenco, D, Hamedian, M, Kawalya, D, Salminen, L, Sequeiros, RB, Koskinen, SK, Pudas, TK, Kajander, S, Niemi, P, Hirvonen, J, Aronen, HJ and Jafaritadi, M (2022) Deep learning accurately classifies elbow joint effusion in adult and pediatric radiographs. Scientific Reports 12(1), 11803.CrossRefGoogle ScholarPubMed
Hunter, T (1995) Protein kinases and phosphatases: The yin and yang of protein phosphorylation and signaling. Cell 80(2), 225236.Google ScholarPubMed
Kandasamy, K, Mohan, SS, Raju, R, Keerthikumar, S, Kumar, GS, Venugopal, AK, Telikicherla, D, Navarro, JD, Mathivanan, S, Pecquet, C, Gollapudi, SK, Tattikota, SG, Mohan, S, Padhukasahasram, H, Subbannayya, Y, Goel, R, Jacob, HK, Zhong, J, Sekhar, R, Nanjappa, V, Balakrishnan, L, Subbaiah, R, Ramachandra, YL, Rahiman, BA, Prasad, TS, Lin, JX, Houtman, JC, Desiderio, S, Renauld, JC, Constantinescu, SN, Ohara, O, Hirano, T, Kubo, M, Singh, S, Khatri, P, Draghici, S, Bader, GD, Sander, C, Leonard, WJ and Pandey, A (2010) NetPath: A public resource of curated signal transduction pathways. Genome Biology 11, R3.Google ScholarPubMed
Kanehisa, M, Goto, S, Sato, Y, Furumichi, M and Tanabe, M (2012) KEGG for integration and interpretation of large-scale molecular data sets. Nucleic Acids Research 40, D109D114.Google ScholarPubMed
Kang, MJ, Ahn, HS, Lee, SY, Yeom, J, Kim, K and Hong, SJ (2022) TGFβ1 and POSTN as biomarkers of postinfectious bronchiolitis obliterans and asthma in children. Pediatric Pulmonology 57(12), 31613164.CrossRefGoogle ScholarPubMed
Kastelein, JJ, Ross, CJ and Hayden, MR (2013) From mutation identification to therapy: Discovery and origins of the first approved gene therapy in the Western world. Human Gene Therapy 24(5), 472478.Google ScholarPubMed
Keam, SJ (2021) Elivaldogene Autotemcel: First approval. Molecular Diagnosis & Therapy 25(6), 803809.CrossRefGoogle ScholarPubMed
Kerrien, S, Aranda, B, Breuza, L, Bridge, A, Broackes-Carter, F, Chen, C, Duesbury, M, Dumousseau, M, Feuermann, M, Hinz, U, Jandrasits, C, Jimenez, RC, Khadake, J, Mahadevan, U, Masson, P, Pedruzzi, I, Pfeiffenberger, E, Porras, P, Raghunath, A, Roechert, B, Orchard, S and Hermjakob, H (2012) The IntAct molecular interaction database in 2012. Nucleic Acids Research 40, D841D846.Google ScholarPubMed
Koledova, E, Tornincasa, V and van Dommelen, P (2020) Analysis of real-world data on growth hormone therapy adherence using a connected injection device. BMC Medical Informatics and Decision Making 20(1), 176.CrossRefGoogle ScholarPubMed
Lalonde, E, Rentas, S, Lin, F, Dulik, MC, Skraban, CM and Spinner, NB (2020) Genomic diagnosis for pediatric disorders: Revolution and evolution. Frontiers in Pediatrics 8, 373.Google ScholarPubMed
Lam, ET, Hastie, A, Lin, C, Ehrlich, D, Das, SK, Austin, MD, Deshpande, P, Cao, H, Nagarajan, N, Xiao, M and Kwok, PY (2012) Genome mapping on nanochannel arrays for structural variation analysis and sequence assembly. Nature Biotechnology 30(8), 771776.Google ScholarPubMed
Langley, RJ and Wong, HR (2017) Early diagnosis of sepsis: Is an integrated omics approach the way forward? Molecular Diagnosis & Therapy 21(5), 525537.CrossRefGoogle ScholarPubMed
Lee, JA, Cho, A, Huang, EN, Xu, Y, Quach, H, Hu, J and Wong, AP (2021) Gene therapy for cystic fibrosis: New tools for precision medicine. Journal of Translational Medicine 19(1), 452.Google ScholarPubMed
Lee, H, Eun, Y, Hwang, JY and Eun, LY (2022) Explainable deep learning algorithm for distinguishing incomplete Kawasaki disease by coronary artery lesions on echocardiographic imaging. Computer Methods and Programs in Biomedicine 223, 106970.Google ScholarPubMed
Lee, J, Gillam, L, Visvanathan, K, Hansford, JR and McCarthy, MC (2021) Clinical utility of precision medicine in pediatric oncology: A systematic review. JCO Precision Oncology 5, 10881102.CrossRefGoogle ScholarPubMed
Lee, JK, Jeong, E, Lee, J, Jung, M, Shin, E, Kim, YH, Lee, K, Jung, I, Kim, D, Kim, S and Kim, JS (2018) Directed evolution of CRISPR-Cas9 to increase its specificity. Nature Communications 9(1), 3048.CrossRefGoogle ScholarPubMed
Leoni, C, Blandino, R, Delogu, AB, De Rosa, G, Onesimo, R, Verusio, V, Marino, MV, Lanza, GA, Rigante, D, Tartaglia, M and Zampino, G (2022) Genotype-cardiac phenotype correlations in a large single-center cohort of patients affected by RASopathies: Clinical implications and literature review. American Journal of Medical Genetics. Part A 188(2), 431445.CrossRefGoogle Scholar
Lestringant, V, Duployez, N, Penther, D, Luquet, I, Derrieux, C, Lutun, A, Preudhomme, C, West, M, Ouled-Haddou, H, Devoldere, C, Marolleau, JP, Garçon, L, Jedraszak, G and Ferret, Y (2021) Optical genome mapping, a promising alternative to gold standard cytogenetic approaches in a series of acute lymphoblastic leukemias. Genes, Chromosomes & Cancer 60(10), 657667.CrossRefGoogle Scholar
Levy, B and Wapner, R (2018) Prenatal diagnosis by chromosomal microarray analysis. Fertility and Sterility 109, 201212.CrossRefGoogle ScholarPubMed
Linzey, JR, Marini, BL, Pasternak, A, Smith, C, Miklja, Z, Zhao, L, Kumar-Sinha, C, Paul, A, Harris, N, Robertson, PL, Hoffman, LM, Chinnaiyan, A, Mody, R and Koschmann, C (2018) Development of the CNS TAP tool for the selection of precision medicine therapies in neuro-oncology. Journal of Neuro-Oncology 137(1), 155169.CrossRefGoogle ScholarPubMed
Lionel, AC, Costain, G, Monfared, N, Walker, S, Reuter, MS, Hosseini, SM, Thiruvahindrapuram, B, Merico, D, Jobling, R, Nalpathamkalam, T, Pellecchia, G, Sung, WWL, Wang, Z, Bikangaga, P, Boelman, C, Carter, MT, Cordeiro, D, Cytrynbaum, C, Dell, SD, Dhir, P, Dowling, JJ, Heon, E, Hewson, S, Hiraki, L, Inbar-Feigenberg, M, Klatt, R, Kronick, J, Laxer, RM, Licht, C, MacDonald, H, Mercimek-Andrews, S, Mendoza-Londono, R, Piscione, T, Schneider, R, Schulze, A, Silverman, E, Siriwardena, K, Snead, OC, Sondheimer, N, Sutherland, J, Vincent, A, Wasserman, JD, Weksberg, R, Shuman, C, Carew, C, Szego, MJ, Hayeems, RZ, Basran, R, Stavropoulos, DJ, Ray, PN, Bowdin, S, Meyn, MS, Cohn, RD, Scherer, SW and Marshall, CR (2018) Improved diagnostic yield compared with targeted gene sequencing panels suggests a role for whole-genome sequencing as a first-tier genetic test. Genetics in Medicine 20(4), 435443.Google ScholarPubMed
Liu, J, Gaj, T, Yang, Y, Wang, N, Shui, S, Kim, S, Kanchiswamy, CN, Kim, JS and Barbas, CF (2015) Efficient delivery of nuclease proteins for genome editing in human stem cells and primary cells. Nature Protocols 10(11), 18421859.CrossRefGoogle ScholarPubMed
Liu, L, Johnson, HL, Cousens, S, Perin, J, Scott, S, Lawn, JE, Rudan, I, Campbell, H, Cibulskis, R, Li, M, Mathers, C and Black, RE (2012) Global, regional, and national causes of child mortality: An updated systematic analysis for 2010 with time trends since 2000. Lancet 379, 21512161.Google ScholarPubMed
Liu, S, Perdew, M, Lithopoulos, A and Rhodes, RE (2021) The feasibility of using Instagram data to predict exercise identity and physical activity levels: Cross-sectional observational study. Journal of Medical Internet Research 23(4), e20954.CrossRefGoogle ScholarPubMed
Mantere, T, Neveling, K, Pebrel-Richard, C, Benoist, M, van der Zande, G, Kater-Baats, E, Baatout, I, van Beek, R, Yammine, T, Oorsprong, M, et al. (2020) Next generation cytogenetics: Genome-imaging enables comprehensive structural variant detection for 100 constitutional chromosomal aberrations in 85 samples. https://www.biorxiv.org/content/10.1101/2020.07.15.205245v1.CrossRefGoogle Scholar
Marks, LJ, Oberg, JA, Pendrick, D, Sireci, AN, Glasser, C, Coval, C, Zylber, RJ, Chung, WK, Pang, J, Turk, AT, Hsiao, SJ, Mansukhani, MM, Glade Bender, JL, Kung, AL and Sulis, ML (2017) Precision medicine in children and young adults with hematologic malignancies and blood disorders: The Columbia University experience. Frontiers in Pediatrics 5, 265.CrossRefGoogle Scholar
Martin Sanchez, F, Gray, K, Bellazzi, R and Lopez-Campos, G (2014) Exposome informatics: Considerations for the design of future biomedical research information systems. Journal of the American Medical Informatics Association 21(3), 386390.CrossRefGoogle ScholarPubMed
Masino, AJ, Harris, MC, Forsyth, D, Ostapenko, S, Srinivasan, L, Bonafide, CP, Balamuth, F, Schmatz, M and Grundmeier, RW (2019) Machine learning models for early sepsis recognition in the neonatal intensive care unit using readily available electronic health record data. PLoS One 14(2), e0212665.CrossRefGoogle ScholarPubMed
Mendell, JR, Al-Zaidy, SA, Rodino-Klapac, LR, et al. (2021) Current clinical applications of in vivo gene therapy with AAVs. Molecular Therapy 29(2), 464488.CrossRefGoogle ScholarPubMed
Meng, L, Pammi, M, Saronwala, A, Magoulas, P, Ghazi, AR, Vetrini, F, Zhang, J, He, W, Dharmadhikari, AV, Qu, C, Ward, P, Braxton, A, Narayanan, S, Ge, X, Tokita, MJ, Santiago-Sim, T, Dai, H, Chiang, T, Smith, H, Azamian, MS, Robak, L, Bostwick, BL, Schaaf, CP, Potocki, L, Scaglia, F, Bacino, CA, Hanchard, NA, Wangler, MF, Scott, D, Brown, C, Hu, J, Belmont, JW, Burrage, LC, Graham, BH, Sutton, VR, Craigen, WJ, Plon, SE, Lupski, JR, Beaudet, AL, Gibbs, RA, Muzny, DM, Miller, MJ, Wang, X, Leduc, MS, Xiao, R, Liu, P, Shaw, C, Walkiewicz, M, Bi, W, Xia, F, Lee, B, Eng, CM, Yang, Y and Lalani, SR (2017) Use of exome sequencing for infants in intensive care units: Ascertainment of severe single-gene disorders and effect on medical management. JAMA Pediatrics 171(12), e173438.CrossRefGoogle ScholarPubMed
Mi, H, Guo, N, Kejariwal, A and Thomas, PD (2007) PANTHER version 6: Protein sequence and function evolution data with expanded representation of biological pathways. Nucleic Acids Research 35, D247D252.CrossRefGoogle ScholarPubMed
Mickiewicz, B, Thompson, GC, Blackwood, J, Jenne, CN, Winston, BW, Vogel, HJ, Joffe, AR; Alberta Sepsis Network. (2015) Development of metabolic and inflammatory mediator biomarker phenotyping for early diagnosis and triage of pediatric sepsis. Critical Care 19(1), 320.Google ScholarPubMed
National Research Council (US) (2009) A New Biology for the 21st Century: Ensuring the United States Leads the Coming Biology Revolution. Washington, DC: National Academies Press.Google Scholar
National Research Council (US) (2011) Toward Precision Medicine: Building a Knowledge Network for Biomedical Research and a New Taxonomy of Disease. Washington, DC: National Academies PressGoogle Scholar
Neveling, K, Mantere, T, Vermeulen, S, Oorsprong, M, van Beek, R, Kater-Baats, E, Pauper, M, van der Zande, G, Smeets, D, Weghuis, DO, Stevens-Kroef, MJPL and Hoischen, A (2021) Next-generation cytogenetics: Comprehensive assessment of 52 hematological malignancy genomes by optical genome mapping. American Journal of Human Genetics 108(8), 14231435.Google ScholarPubMed
Nijman, RG, Oostenbrink, R, Moll, HA, Casals-Pascual, C, von Both, U, Cunnington, A, De, T, Eleftheriou, I, Emonts, M, Fink, C, van der Flier, M, de Groot, R, Kaforou, M, Kohlmaier, B, Kuijpers, TW, Lim, E, Maconochie, IK, Paulus, S, Martinon-Torres, F, Pokorn, M, Romaine, ST, Calle, IR, Schlapbach, LJ, Smit, FJ, Tsolia, M, Usuf, E, Wright, VJ, Yeung, S, Zavadska, D, Zenz, W, Levin, M, Herberg, JA, Carrol, ED; PERFORM consortium (Personalized Risk assessment in febrile children to optimize Real-life Management across the European Union). (2021) A novel framework for phenotyping children with suspected or confirmed infection for future biomarker studies. Frontiers in Pediatrics 9, 688272.CrossRefGoogle ScholarPubMed
Ó Cathaoir, K (2021) The invisible child of personalized medicine. Journal of Law and the Biosciences 8(2), lsab029.CrossRefGoogle ScholarPubMed
Otero, BA, Poukalov, K, Hildebrandt, RP, Thornton, CA, Jinnai, K, Fujimura, H, Kimura, T, Hagerman, KA, Sampson, JB, Day, JW and Wang, ET (2021) Transcriptome alterations in myotonic dystrophy frontal cortex. Cell Reports 34, 108634.CrossRefGoogle ScholarPubMed
Padash, S, Mohebbian, MR, Adams, SJ, Henderson, RDE and Babyn, P (2022) Pediatric chest radiograph interpretation: How far has artificial intelligence come? A systematic literature review. Pediatric Radiology 52(8), 15681580.CrossRefGoogle ScholarPubMed
Patel, SJ, Chamberlain, DB and Chamberlain, JM (2018) A machine learning approach to predicting need for hospitalization for pediatric asthma exacerbation at the time of emergency department triage. Academic Emergency Medicine 25(12), 14631470.CrossRefGoogle ScholarPubMed
Pavel, AM, Rennie, JM, de Vries, LS, Blennow, M, Foran, A, Shah, DK, Pressler, RM, Kapellou, O, Dempsey, EM, Mathieson, SR, Pavlidis, E, van Huffelen, AC, Livingstone, V, Toet, MC, Weeke, LC, Finder, M, Mitra, S, Murray, DM, Marnane, WP and Boylan, GB (2020) A machine-learning algorithm for neonatal seizure recognition: A multicentre, randomised, controlled trial. The Lancet Child & Adolescent Health 4(10), 740749.CrossRefGoogle ScholarPubMed
Pennisi, I, Moniri, A, Miscourides, N, Miglietta, L, Moser, N, Habgood-Coote, D, Herberg, JA, Levin, M, Kaforou, M, Rodriguez-Manzano, J and Georgiou, P (2022) Discrimination of bacterial and viral infection using host-RNA signatures integrated in a lab-on-chip platform. Biosens Bioelectron. 216:114633.Google Scholar
Pincez, T, Clément, N, Lapouble, E, Pierron, G, Kamal, M, Bieche, I, Bernard, V, Fréneaux, P, Michon, J, Orbach, D, Aerts, I, Pacquement, H, Bourdeaut, F, Jiménez, I, Thébaud, E, Oudot, C, Vérité, C, Taque, S, Owens, C, Doz, F, Le Tourneau, C, Delattre, O and Schleiermacher, G (2017) Feasibility and clinical integration of molecular profiling for target identification in pediatric solid tumors. Pediatric Blood & Cancer 64, e26365.CrossRefGoogle ScholarPubMed
Posey, JE, Harel, T, Liu, P, Rosenfeld, JA, James, RA, Coban Akdemir, ZH, Walkiewicz, M, Bi, W, Xiao, R, Ding, Y, Xia, F, Beaudet, AL, Muzny, DM, Gibbs, RA, Boerwinkle, E, Eng, CM, Sutton, VR, Shaw, CA, Plon, SE, Yang, Y and Lupski, JR (2017) Resolution of disease phenotypes resulting from multilocus genomic variation. The New England Journal of Medicine 376(1), 2131.CrossRefGoogle ScholarPubMed
Qasim, W, Gaspar, HB and Thrasher, AJ (2007) Update on clinical gene therapy in childhood. Archives of Disease in Childhood 92(11), 10281031.CrossRefGoogle ScholarPubMed
Rack, K, De Bie, J, Ameye, G, Gielen, O, Demeyer, S, Cools, J, De Keersmaecker, K, Vermeesch, JR, Maertens, J, Segers, H and Michaux, L and Dewaele, B (2022) Optimizing the diagnostic workflow for acute lymphoblastic leukemia by optical genome mapping. American Journal of Hematology 97(5), 548561.CrossRefGoogle ScholarPubMed
Raita, Y, Camargo, CA Jr, Macias, CG, Mansbach, JM, Piedra, PA, Porter, SC, Teach, SJ and Hasegawa, K (2020) Machine learning-based prediction of acute severity in infants hospitalized for bronchiolitis: A multicenter prospective study. Scientific Reports 10(1), 10979.Google ScholarPubMed
Rauen, KA (2013) The RASopathies. Annual Review of Genomics and Human Genetics 14, 355369.Google ScholarPubMed
Reece, AG and Danforth, CM (2017) Instagram photos reveal predictive markers of depression. EPJ Data Science 6, 112.Google Scholar
Reece, AG, Reagan, AJ, Lix, KLM, Dodds, PS, Danforth, CM and Langer, EJ (2017) Forecasting the onset and course of mental illness with twitter data. Scientific Reports 7(1), 13006.Google ScholarPubMed
Riller, Q and Rieux-Laucat, F (2021) RASopathies: From germline mutations to somatic and multigenic diseases. Biomedical Journal 44(4), 422432.Google ScholarPubMed
Rivat, C, Santilli, G, Gaspar, HB and Thrasher, AJ (2012) Gene therapy for primary immunodeficiencies. Human Gene Therapy 23(7), 668675.Google ScholarPubMed
Sahajpal, NS, Barseghyan, H, Kolhe, R, Hastie, A and Chaubey, A (2021) Optical genome mapping as a next-generation cytogenomic tool for detection of structural and copy number variations for prenatal genomic analyses. Genes (Basel) 12(3), 398.Google ScholarPubMed
Salsman, J and Dellaire, G (2017) Precision genome editing in the CRISPR era. Biochemistry and Cell Biology 95(2), 187201.Google ScholarPubMed
Sankilampi, U, Saari, A, Laine, T, Miettinen, PJ and Dunkel, L (2013) Use of electronic health records for automated screening of growth disorders in primary care. Journal of the American Medical Association 310(10), 10711072.CrossRefGoogle ScholarPubMed
Schaefer, CF, Anthony, K, Krupa, S, Buchoff, J, Day, M, Hannay, T and Buetow, KH (2009) PID: The pathway interaction database. Nucleic Acids Research 37, D674D679.CrossRefGoogle ScholarPubMed
Schmidt, A, Forne, I and Imhof, A (2014) Bioinformatic analysis of proteomics data. BMC Systems Biology 8(Suppl 2), S3.Google ScholarPubMed
Shieh, JT, Penon-Portmann, M, Wong, KH, Levy-Sakin, M, Verghese, M, Slavotinek, A, Gallagher, RC, Mendelsohn, BA, Tenney, J, Beleford, D, Perry, H, Chow, SK, Sharo, AG, Brenner, SE, Qi, Z, Yu, J, Klein, OD, Martin, D, Kwok, PY and Boffelli, D (2020) Application of full-genome analysis to diagnose rare monogenic disorders. NPJ Genom Med. 6(1):77.CrossRefGoogle Scholar
Sillence, DO, Senn, A and Danks, DM (1979) Genetic heterogeneity in osteogenesis imperfecta. Journal of Medical Genetics 16, 101116.CrossRefGoogle ScholarPubMed
Spataru, A, Quarteroni, S, Arnaud, L, van Dommelen, P, Koledova, E and Le Masne, Q (2021) High engagement of patients monitored by a digital health ecosystem indicates significant improvements of key r-hGH treatment metrics. Studies in Health Technology and Informatics 281, 829833.Google ScholarPubMed
Spataru, A, van Dommelen, P, Arnaud, L, Le Masne, Q, Quarteroni, S and Koledova, E (2022) Use of machine learning to identify patients at risk of sub-optimal adherence: Study based on real-world data from 10,929 children using a connected auto-injector device. BMC Medical Informatics and Decision Making 22(1), 179.CrossRefGoogle ScholarPubMed
Stark, Z and Ellard, S (2022) Rapid genomic testing for critically ill children: Time to become standard of care? European Journal of Human Genetics 30(2), 142149.CrossRefGoogle ScholarPubMed
Stavropoulos, DJ, Merico, D, Jobling, R, Bowdin, S, Monfared, N, Thiruvahindrapuram, B, Nalpathamkalam, T, Pellecchia, G, Yuen, RKC, Szego, MJ, Hayeems, RZ, Shaul, RZ, Brudno, M, Girdea, M, Frey, B, Alipanahi, B, Ahmed, S, Babul-Hirji, R, Porras, RB, Carter, MT, Chad, L, Chaudhry, A, Chitayat, D, Doust, SJ, Cytrynbaum, C, Dupuis, L, Ejaz, R, Fishman, L, Guerin, A, Hashemi, B, Helal, M, Hewson, S, Inbar-Feigenberg, M, Kannu, P, Karp, N, Kim, R, Kronick, J, Liston, E, MacDonald, H, Mercimek-Mahmutoglu, S, Mendoza-Londono, R, Nasr, E, Nimmo, G, Parkinson, N, Quercia, N, Raiman, J, Roifman, M, Schulze, A, Shugar, A, Shuman, C, Sinajon, P, Siriwardena, K, Weksberg, R, Yoon, G, Carew, C, Erickson, R, Leach, RA, Klein, R, Ray, PN, Meyn, MS, Scherer, SW, Cohn, RD and Marshall, CR (2016) Whole genome sequencing expands diagnostic utility and improves clinical management in pediatric medicine. NPJ Genomic Medicine 1, 15012.CrossRefGoogle ScholarPubMed
Stratmann, S, Vesterlund, M, Umer, HM, Eshtad, S, Skaftason, A, Herlin, MK, Sundström, C, Eriksson, A, Höglund, M, Palle, J, Abrahamsson, J, Jahnukainen, K, Munthe-Kaas, MC, Zeller, B, Tamm, KP, Lindskog, C, Cavelier, L, Lehtiö, J and Holmfeldt, L (2022) Proteogenomic analysis of acute myeloid leukemia associates relapsed disease with reprogrammed energy metabolism both in adults and children. Leukemia 26.Google Scholar
Streekstra, EJ, Russel, FGM, van de Steeg, E and de Wildt, SN (2021) Application of proteomics to understand maturation of drug metabolizing enzymes and transporters for the optimization of pediatric drug therapy. Drug Discovery Today: Technologies 39, 3148,CrossRefGoogle ScholarPubMed
Su, PH, Malik, S, Jheeta, A, Lin, YF, Su, SH, Koledova, E and Graham, S (2022) Investigating the impact of the TUITEK® patient support Programme, designed to support caregivers of children prescribed recombinant human Growth hormone treatment in Taiwan. Frontiers in Endocrinology 13, 897956.Google ScholarPubMed
Titano, JJ, Badgeley, M, Schefflein, J, Pain, M, Su, A, Cai, M, Swinburne, N, Zech, J, Kim, J, Bederson, J, Mocco, J, Drayer, B, Lehar, J, Cho, S, Costa, A and Oermann, EK (2018) Automated deep-neural-network surveillance of cranial images for acute neurologic events. Nature Medicine 24(9), 13371341.CrossRefGoogle ScholarPubMed
Trincianti, C, Meleca, V, La Porta, E, Bruschi, M, Candiano, G, Garbarino, A, Kajana, X, Preda, A, Lugani, F, Ghiggeri, GM, Angeletti, A, Esposito, P and Verrina, E (2022) Proteomics and extracellular vesicles as novel biomarker sources in peritoneal dialysis in children. International Journal of Molecular Sciences 23(10), 5655.Google ScholarPubMed
Uddin, F, Rudin, CM and Sen, T (2020) CRISPR gene therapy: Applications, limitations, and implications for the future. Frontiers in Oncology 10, 1387.CrossRefGoogle ScholarPubMed
van Dommelen, P, Koledova, E and Wit, JM (2018) Effect of adherence to growth hormone treatment on 0-2 year catch-up growth in children with growth hormone deficiency. PLoS One 13(10), e0206009.CrossRefGoogle ScholarPubMed
van Groen, BD, Allegaert, K, Tibboel, D and de Wildt, SN (2022) Innovative approaches and recent advances in the study of ontogeny of drug metabolism and transport. British Journal of Clinical Pharmacology 88(10), 42854296.Google Scholar
Vo, KT, Parsons, DW and Seibel, NL (2020) Precision medicine in pediatric oncology. Surgical Oncology Clinics of North America 29(1), 6372.Google ScholarPubMed
Vrijheid, M (2014) The exposome: A new paradigm to study the impact of environment on health. Thorax 69(9), 876878.CrossRefGoogle Scholar
Wang, E, Cho, WCS, Wong, SCC and Liu, S (2017) Disease biomarkers for precision medicine: Challenges and future opportunities. Genomics, Proteomics & Bioinformatics 15(2), 5758.CrossRefGoogle ScholarPubMed
Wild, CP (2012) The exposome: From concept to utility. International Journal of Epidemiology 41(1), 2432.CrossRefGoogle ScholarPubMed
Wilkins, MR, Sanchez, JC, Gooley, AA, Appel, RD, Humphery-Smith, I, Hochstrasser, DF and Williams, KL (1996) Progress with proteome projects: Why all proteins expressed by a genome should be identified and how to do it. Biotechnology & Genetic Engineering Reviews 13, 1950.Google ScholarPubMed
Williams, AM, Liu, Y, Regner, KR, Jotterand, F, Liu, P and Liang, M (2018) Artificial intelligence, physiological genomics, and precision medicine. Physiological Genomics 50(4), 237243.CrossRefGoogle ScholarPubMed
Wong, HR, Atkinson, SJ, Cvijanovich, NZ, et al. (2016) Combining prognostic and predictive enrichment strategies to identify children with septic shock responsive to corticosteroids. Critical Care Medicine 44(10), e1000e1003.Google ScholarPubMed
Wong, HR, Atkinson, SJ, Cvijanovich, NZ, Anas, N, Allen, GL, Thomas, NJ, Bigham, MT, Weiss, SL, Fitzgerald, JC, Checchia, PA, Meyer, K, Quasney, M, Hall, M, Gedeit, R, Freishtat, RJ, Nowak, J, Raj, SS, Gertz, S and Lindsell, CJ (2011) Validation of a gene expression-based subclassification strategy for pediatric septic shock. Critical Care Medicine 39(11), 25112517.CrossRefGoogle ScholarPubMed
Wong, HR, Cvijanovich, NZ, Anas, N, Allen, GL, Thomas, NJ, Bigham, MT, Weiss, SL, Fitzgerald, J, Checchia, PA, Meyer, K, Shanley, TP, Quasney, M, Hall, M, Gedeit, R, Freishtat, RJ, Nowak, J, Shekhar, RS, Gertz, S, Dawson, E, Howard, K, Harmon, K, Beckman, E, Frank, E and Lindsell, CJ (2015) Developing a clinically feasible personalized medicine approach to pediatric septic shock. American Journal of Respiratory and Critical Care Medicine 191(3), 309315.Google ScholarPubMed
Wong, HR, Cvijanovich, NZ, Anas, N, Allen, GL, Thomas, NJ, Bigham, MT, Weiss, SL, Fitzgerald, JC, Checchia, PA, Meyer, K, Quasney, M, Hall, M, Gedeit, R, Freishtat, RJ, Nowak, J, Raj, SS, Gertz, S, Grunwell, JR and Lindsell, CJ (2017) Improved risk stratification in pediatric septic shock using both protein and mRNA biomarkers. PERSEVERE-XP. American Journal of Respiratory and Critical Care 196(4), 494501.CrossRefGoogle ScholarPubMed
Wong, HR, Cvijanovich, N, Lin, R, et al. (2009) Identification of pediatric septic shock subclasses based on genome-wide expression profiling. BMC Medicine 7, 34.CrossRefGoogle ScholarPubMed
Wong, HR, Shanley, TP, Sakthivel, B, et al. (2007) Genome-level expression profiles in pediatric septic shock indicate a role for altered zinc homeostasis in poor outcome. Physiological Genomics 30(2), 146155.CrossRefGoogle ScholarPubMed
Wong, HR, Sweeney, TE and Lindsell, CJ (2017) Simplification of a septic shock endotyping strategy for clinical application. American Journal of Respiratory and Critical Care Medicine 195(2), 263265.CrossRefGoogle ScholarPubMed
Ylä-Herttuala, S (2012) Endgame: Glybera finally recommended for approval as the first gene therapy drug in the European union. Molecular Therapy 20(10):18311832.CrossRefGoogle ScholarPubMed
Zaripova, AR and Khusainova, RI (2020) Modern classification and molecular-genetic aspects of osteogenesis imperfecta. Vavilovskii Zhurnal Genetiki i Selektsii 24(2), 219227.Google ScholarPubMed
Zhu, J, Garrigues, L, Van den Toorn, H, Stahl, B and Heck, AJR (2019) Discovery and quantification of nonhuman proteins in human Milk. Journal of Proteome Research 18(1), 225238.Google ScholarPubMed
Zuercher, M, Ufkes, S, Erdman, L, Slorach, C, Mertens, L and Taylor, K (2022) Retraining an artificial intelligence algorithm to calculate left ventricular ejection fraction in pediatrics. Journal of Cardiothoracic and Vascular Anesthesia 36(9), 36103616.CrossRefGoogle ScholarPubMed

Author comment: Precision diagnostics in children — R0/PR1

Comments

Dear Editor

Thank you for inviting me to submit my manuscript on precision diagnostics in paediatrics. I trust that you will find my manuscript an accurate reflection on the current advances in the sciences

Yours Sincerely

Paul Dimitri

Review: Precision diagnostics in children — R0/PR2

Conflict of interest statement

Reviewer declares none.

Comments

Comments to Author: 1- English: The manuscript could benefit from editing for grammar, missing words, and subject-verb agreement, etc. It is recommended that authors delete irrelevant "general" phrases and sentences, repeated and unneeded words. They should use short sentences. Also, some Introductory sentences are irrelevant or are not needed. There are also typos in the manuscript.

2- Abbreviations: All abbreviations should be revised and defined at their first use.

3- Abstract: The abstract is very general and does not tackle pediatric population or diseases at all. The main purpose of the review is not clearly defined in the abstract. What is actually mentioned are all known facts that precision medicine and patient centered care is the future.

4- Introduction: “participatory component of P4 medicine” it is confusing what is meant by P4? Is it the same as 4Ps?

5- Introduction: “data capture from an early age will rely on empowering children and young people to contribute their data, motivating them to maintain this activity and individual trust that data will be utilised appropriately and stored secure.” What exactly do those data encompass? Please elaborate and explain how individuals can contribute their data?

6- Introduction: talking about sharing personal health data from early childhood to adulthood, isn’t this breach of patient privacy? What protects this privacy?

7- Introduction: there is no mention of a single word related to pediatrics in the introduction. The author should have a good flow of ideas where he jumps from one section to another using liaisons.

8- Section “Disease reclassification and predicting future disease in children”: in this section, I do not feel the authors provided any interesting finding that could be beneficial for readers. It is well known that reclassification of many diseases and cancers based on molecular signatures is a reality we are living, such as brain tumors.

9- Under “Developing biomarkers and targeted therapies to treat childhood diseases”: the author also tackled proteomics, predictive biomarkers, and precision diagnostics for targeted drug therapies, from a very broad perspective. In the section proteomics, the author just defined what proteomic medicine is about, without giving examples from the pediatric world. Similarly, under predictive biomarkers, not a single example is provided.

Recommendation: Precision diagnostics in children — R0/PR3

Comments

Comments to Author: I agree that the details that Reviewer #1 points out all do have some truth to them, however I think they are in general a little overstated in their severity and hence I believe the article can be ready to publish with only some revisions satisfying the spirit behind Reviewer #1 comments and my additional review points below. Overall, the article is a highly relevant and beneficial contribution to an important aspect of precision medicine, that of how it impacts the health of patients early in life.

• Example: yes, the author interchanges P4 and 4P, most often using what I believe the non-standard abbreviation (and as used in the cited references). However, they do define their abbreviation at the first use. Simple to fix. Otherwise, I don’t see an abnormal rate of grammatical errors.

o Page 4: single-quotes mis-spaced in “individual data profile”

o Page 5: “revolutionise our ability to modify”

o Page 9: no hyphen for in vitro or in vivo (ex vivo on p11, “in turn” on 16)

• I agree that the author will make this good article even better by describing some additional examples as referred to by Reviewer #1 (especially the proteomics section, since concrete examples are provided in the subsequent section), but to be fair, this is a rather broad-reaching review and I think it is thoroughly blanketed in appropriate citations even if not fully discussed.

• I believe the article would do well to discuss in a bit further detail the inherent “problem” of germline genetic disease, where there may not be a cure/treatment…and the primary benefit is either to prepare families for appropriate palliative care or to rule out other more actionable diseases. This is a common conundrum/issue in the field, so addressing it head on in more depth than currently mentioned in the reclassification section would be good.

• I’m slightly concerned about the absence of specific/depth of coverage of next generation cytogenetics methods (eg., CMA or optical mapping), but perhaps the author has good reason to omit this contribution to the completeness of this review (as I recognize the breadth of the review inherently limits how much depth can be covered).

• I feel the author’s pointing out the need for “legal and governance structures” is sufficient counterbalance to address Reviewer #1’s concerns in this area. Perhaps the concern hints at a benefit of pointing out that the solutions will likely vary based on localised laws and culture.

Decision: Precision diagnostics in children — R0/PR4

Comments

No accompanying comment.

Author comment: Precision diagnostics in children — R1/PR5

Comments

To the Editor-in-Chief

I have sent you the revised version of my manuscript for publication, after addressing the comments and suggestion made by the handling Editor and Reviewer. I trust that my amendments provide a satisfactory response to proceed to publication

Kind regards

Paul Dimitri

Recommendation: Precision diagnostics in children — R1/PR6

Comments

Comments to Author: This manuscript, and especially the additional examples included in this revision, is a very nice review of the state of and important benefits of precision medicine in the context of childhood disease.

Decision: Precision diagnostics in children — R1/PR7

Comments

No accompanying comment.