Respiratory metagenomics: route to routine service

INTRODUCTION

Clinicians review many sources of information when assessing patients with an acute infectious disease, which for the purposes of this review is on the adult or paediatric intensive care unit (ICU) with a suspected severe lower respiratory tract infection (LRTI). There are multiple viral, bacterial, and fungal pathogens, some with potential for transmission to family, recent community contacts and now healthcare staff, and there may be antimicrobial resistance which complicates treatment choices, particularly if the LRTI was acquired in hospital. There is also the rare but not discountable possibility of novel pathogens including with pandemic potential as happened with Middle East Respiratory Syndrome Coronavirus (MERS-CoV) on our own ICU in 2012 [1] and South Korea in 2015 [2] and with Severe Acute Respiratory Syndrome (SARS)-CoV-2 in 2019 in China [3].

Respiratory and other samples are tested, each for a selected range of pathogens and often in different laboratories that provide results back from within an hour, to a few hours, the next day or many days, and weeks later. Data from biochemistry, haematology, imaging, history, examination, and physiological parameters are available that day, but the identity and antimicrobial resistance (AMR) of causative pathogens are not. Consequently, treatment, infection control and public health decisions are made empirically at different times based on the likelihood and consequence of different pathogens. A total of 25–47% of community LRTI cases remain undiagnosed after all conventional testing [4], and presumably more if certain tests are not requested. Clinicians in every acute hospital in the world deal with this uncertainty every day [5▪]. 

FB1Box 1:

no caption available

THE RESPIRATORY METAGENOMICS PROPOSITION

The clinical metagenomics proposition in an acute care setting is to sequence nucleic acid from all organisms in a patient sample, to provide all required pathogen information ideally on the day of sample collection. Rapid turnaround time is important but shared by other molecular tests, however it is the comprehensive pathogen-agnostic capability that is the unique aspect setting metagenomics apart from all other tests. The ability to identify conventional agents and their AMR profile, alongside the unexpected, unrequested, hard to detect or novel pathogens, and therefore conversely a more dependable “rule-out” result. Application to respiratory infection has potential for greatest impact, with data from a single test used for patient treatment, antimicrobial stewardship, hospital infection control, public health surveillance and pandemic preparedness [6,7].

This opportunity was clearly articulated before the pandemic along with equally matched technical and implementation challenges [8,9], such that progress towards routine service was considered modest. The pandemic changed this balance for many reasons and expedited movement of respiratory metagenomics (RMg) along the translational research pathway (Fig. 1). RMg provided the full SARS-CoV-2 sequence from the first coronavirus disease 2019 (COVID-19) patients [3] and as the pandemic progressed new sequencing capability was placed in infectious diseases laboratories worldwide for SARS-CoV-2 genomic epidemiology. As clinical and academic teams came together through the unifying pandemic imperative [10], the translational research timeline for pathogen sequencing shortened. Diagnostic accuracy studies are beginning to demonstrate satisfactory performance with pathogen detection [11▪,12▪] and there are innovative approaches to detecting AMR elements using nanopore sequencing with cas9 enrichment [13▪] or predicting AMR using machine learning [14]. There is also increasing focus on clinical laboratory quality requirements including validation, reproducibility, setting limits of detection, and producing positive and negative internal controls and externally distributed reference materials for quality assurance [15]. Rapid workflows have been developed [16], evaluated in research setting during the pandemics [17] and taken through into a service setting (Charalampous – manuscript in preparation) using established hospital quality improvement governance processes [18▪]. Sequencing companies adapted their offer to infectious diseases, recognising the greater turnaround time imperative compared with human genomic sequencing. Illumina have smaller iSeq 100 and MiniSeq platforms while Oxford Nanopore Technologies have developed the portable Minion with single sample adaptors (Flongle), providing results from end-to-end workflows in <6 h, considered particularly suited for further evaluation in a clinical setting [19]. Detailed characteristics of sequencing platforms have been recently reviewed and not covered here [15,20,21].

F1FIGURE 1:

Integrating contribution from different stakeholders to the respiratory metagenomics route to routine service. This is an illustrative perspective from a clinical academic laboratory. Stakeholders will use different terminologies and have different views on ordering and prioritisation of activities which are not necessarily sequential. The core principle is close iterative engagement between each workstream. (a) Industry workstream involves technology development, ISO9001 or ISO13485 accreditation of their products and submission to regulators. (b) Basic, translational and clinical scientist workstream. Involves technology co-development, workflow refinement to optimise performance characteristics, evaluation in small laboratory studies using clinical samples, pilot local service evaluation of internally developed workflows under clinical governance framework, through to ISO15189 accreditation as a laboratory developed test generating real world evidence and involvement in large clinical trials (c) Clinical and service user workstream of all hospital based stakeholders including service providers (microbiology and infectious diseases), infection specialty service users (infection control & antimicrobial stewardship), clinical users (respiratory and intensive care physicians), service enables (IT) and internal decision makers and funders (hospital management). Three main stages of close inter-workstream engagement are represented by vertical filled arrows as (1) Proof-of-concept stage (circa 50–100 sample studies) that a workflow can be safely and reliably performed delivering results that have potential for clinically utility in a required timeframe. This gives support for each workstream to progress to (2) Proof-of-value (circa 500–1000 sample studies) involving early clinical evaluation generating real-world evidence and assessment as a locally developed and accredited test with defined performance characteristics, which informs 3) Proof-of-implementation requiring investment from industry in final product development and regulatory approval, by research funders to support large multisite clinical trials to demonstrate clinical utility and cost-effectiveness and, by healthcare systems implementing infrastructure, producing business cases for using tests, training staff and re-organising clinical pathways to realise benefits. Dotted red line indicates approximate current position.

With these technical advances have come increasing case reports and series identifying unusual or unexpected pathogens [22–28], comparative evaluations [29–31] and literature reviews [32▪,33] demonstrating the value of RMg in a real-world adult and paediatric setting. Many were retrospective or observational, but some informed treatment in real time [34] and the protocol for a randomised multicentre clinical trial has been published [35▪]. RMg consistently increases identification of causative pathogens, in one paediatric ICU study including noninfective control patients up from 67% with all conventional testing to 92% [36▪]. There also remains the need to distinguish infection from colonisation which applies to all microbiological tests. This is particularly relevant for opportunistic infections that require additional investigations and prolonged treatment. RMg provides rapid identification of Pneumocystis jirovecci in non-human immunodeficiency virus immunosuppressed patients [37,38], but could lead to unnecessary investigation and overtreatment where significance is uncertain, as might organisms not conventionally detected in the respiratory tract such as Trophorema whipplei[39]. These will be important factors to assess in clinical and health-economic studies. Inclusion of the host inflammatory response signature alongside relative abundance of putative pathogens will be a helpful addition [40].

A ROUTE TO IMPLEMENTATION IN THE HEALTHCARE SYSTEM

Given these recent developments this review considers an end-state where every patient having a deep respiratory sample submitted to the diagnostic laboratory has first-line metagenomic testing as standard of care. Culture and targeted molecular tests continue for selected cases or as reflex tests, rather than the other way round, which is the current generally accepted position [21,41]. Targeted testing in selected institutions is an inequitable end-state only partially delivering the benefit of RMg and will be hard to sustain in an acute care pathway dependent on heightened awareness by clinicians and scientists diverting exceptional samples away from the routine sample pathway. This review therefore looks ahead to routine adoption, to consider requirements and potential benefits for clinical teams, laboratory networks and the wider healthcare system (Fig. 2).

F2FIGURE 2:

Integrating multidisciplinary working and data flows across the infectious diseases network to realise benefits of respiratory metagenomics. Stakeholder roles and connections currently exist, but information transfer is incomplete and staggered over days, weeks or months. Data is pushed or pulled through different channels in different formats (individual paper or electronic patient reports, spreadsheets, pdf files, phone calls or e mails) with variable interpretation or prioritisation. Transfer often too slow and laborious to meet clinically required time-points so opportunities for prevention, diagnosis or treatment are missed. Lack of common real-time data-visibility frustrates shared learning and prioritisation, and keeps infection professions in silos working cross department, organisation and funding boundaries. A single pathogen genetic data-output simultaneously distributed across the infection network, would support shared real-time assessment and prioritisation. Each professional group identifies components of the common dataset required for continuous service improvement in their domain but with visibility across the network. (a) Standardised sample collection (e.g., BAL or mini-BAL) and targets for laboratory transfer times with sample tracking. (b) includes the [i] Targeted amplicon, pure isolate or metagenomic sequencing from molecular laboratory section or bench. [ii] Automated bioinformatic analysis and interpretation, with [iii] clinical and scientific oversight of unexpected or concerning results to correlate with clinical presentation and alert infection network [iv] comprehensive reports to clinicians during the initial diagnostic evaluation phase. (c) daily infectious diseases or clinical microbiology bed-side or virtual rounds enhanced by including (d) antimicrobial pharmacy and (e) infection control representation. (f) Stored pathogen sequence data, with or without metadata from the electronic health record, improves either i) individual reports potentially with machine learning approaches integrated into real-time report generation (green box) or ii) as a repository for off-line analysis and export. (g) Real-time or intermittent sequence data transfer outside the primary care organisation with information governance approvals (red dashed line) for pathogen surveillance and (h) after anonymisation to academic groups with research governance approval to address knowledge gaps in all aspects of the new infectious diseases framework. Outputs from (g) and (h) fed back to the clinical team.

Clinical input to refining metagenomic workflows

Clinical input is needed to define user requirements to develop clear target product profiles that will inform late-stage methodology refinement [42]. It is not straightforward predicting characteristics of a novel molecular test that will ensure uptake into routine service, even by future users themselves. It requires iterative input that can be informed by experience using early prototypes in a pilot setting [18▪]. Methods differ in their efficiency extracting nucleic acid from different organisms, given the wide variation in structure and physico-chemical properties of bacterial types, including mycobacteria and those with or without a cell wall, fungi, deoxyribonucleic acid (DNA) or ribonucleic acid (RNA) viruses, protozoa, and other parasites. Some workflows will also retain free DNA from dead organisms, which may or may not be considered clinically useful [43]. Input is required setting clinically significant thresholds for reporting organisms, some of which may differ. For example, thresholds for reporting DNA sequence from a Gram-negative bacteria in a patient with suspected ventilator associated pneumonia could be set at or below the culture threshold. Conversely, no sensitivity limit would be imposed for obligate respiratory pathogens such as Mycobacterium tuberculosis or Chlamydia psittacci. Indeed, targeted approaches for some respiratory pathogens such as M. tuberculosis when their presence is specifically sought [44], which in principle could be incorporated into the metagenomics assay rather than separately. Limitations of different RMg workflows will impact on safe result interpretation, particularly when considering antimicrobial de-escalation based on no (significant) organisms reported.

The content and time of report production needs consideration. Accurate organism identification is possible from sequencing <0.1% of the genome after a few minutes so there may be benefit producing organism profile or “No (significant) organisms detected” reports first [45]. Alternatively, clinicians may prefer waiting for a single report a few hours later that includes presence of specific resistance or virulence genes and predictions for phenotypic susceptibilities. Typing reports from full genome sequence may come later with more detail or full sequence output to infection control, public health and research groups under research ethics framework. These are important considerations because while technological improvement continues apace, clinical workflows need to focus on relevant actionable information, rather than providing everything achievable at the technology limits. Storing large amounts of organism sequence in clinical systems would not be cost-effective, although potentially attractive for academic or surveillance purposes.

Clinical service and hospital operational requirements

Improving diagnostic pathways will require improvement to the pre-analytic and post-analytic stages in addition to the testing methodology itself. The UK standards for microbiology investigations for blood cultures has set new targets for time to analysis of <4 h alongside bigger blood volumes to increase sensitivity [46]. Similar standards for respiratory samples make no comment on time to analysis other than recording if the sample is not processed on day of collection [47]. International guidelines for hospital and community acquired pneumonia don’t specify turnaround-time either and have different views on quality of sample collection between directed bronchoalveolar lavage samples versus tracheal aspirate [48–50]. This probably reflects the clear link between positive blood culture, rapid sepsis treatment and outcomes [51] that is hard to assess for LRTI when culture is the diagnostic method. RMg can prompt much needed standardisation of turnaround times and quality for the respiratory diagnostic pathway, including for example specifying BALs or less invasive but standardised mini-BAL sampling [52], rather than accepting the heterogeneity associated with tracheal aspirates.

Clinical reporting will need re-evaluation with turnaround times for validation or auto-release rather than batch reporting. At least initially, interpretation may be needed at the bed-side or at daily hybrid MDTs with clinical microbiology, infectious diseases, radiologists, antimicrobial pharmacists and intensivists present. This will be an important opportunity to achieve better consensus about aetiology and diagnosis of LRTI, particularly ventilator-associated pneumonia (VAP), which has suffered from longstanding uncertainty and controversy [53,54] and drives most antimicrobial prescribing on ICU. Scoring systems have attempted to improve VAP diagnosis but are not recommended in guidelines [49], and clinical assessment without supportive microbiology contributes to pneumonia being frequently mis-diagnosed across the emergency pathway [55]. Clinical experience gained by the MDT correlating RMg results with mild or severe infection and noninfective conditions [36▪], should improve the understanding, diagnosis and treatment of LRTIs, including the impact of antibiotics on the resistome linked with treatment duration and antimicrobial stewardship decisions [56]. Over time accumulated datasets can be used to develop automated interpretation and decision support, including through machine learning linking metagenomic patterns with diagnosis, severity, treatment, and outcomes

Quantifying and generating evidence of clinical and health-economic benefit

Providing evidence of combined clinical and health-economic benefits at a system level will be an important driver for RMg adoption and re-imbursement but will likely come from a range of sources and trials that capture the different benefits. They will need to assess (1) Immediate patient benefit, commencing appropriate antimicrobials when organisms are identified predicted to have intrinsic or acquired resistance to empiric therapy. The frequency will depend on prevalence of endemic AMR, local guidelines and individual risk assessment made by attending clinician. (2) AMR benefit, targeting or stopping antimicrobials by identifying organisms predicted to be susceptible to a narrower antimicrobial spectrum or reporting no organisms. Almost all antimicrobial prescribing includes some retrospectively definable over-treatment, so measures of safe achievable reduction are required for clinical trials. (3) Wider patient and organisational benefit through faster identification of organisms with infection control significance prompting interventions to prevent transmission or further cases. (4) Public health benefit, identifying organisms or variants with wider public health significance including increasing community epidemiological trends, new or re-emerging infections.

These benefits will be captured in target product profiles [42] that specify analytical performance alongside clinical validity, utility and cost-effectiveness. Analytical performance studies are relatively straightforward, whereas randomised controlled trials demonstrating clinical endpoints and cost-effectiveness are recognised to be prohibitively expensive for diagnostic tests. They are also conceptually complex because new molecular tests provide information informing individual decisions that are dependent on human beliefs and behaviours [57], as well as the organisations infrastructure and resources to respond. Evidence may need to include real world data [58], virtual patient trial design [59] and proposals for more efficient trial methodologies that assess the realisable clinical benefit of CMg tests will be welcomed.

Finally, there is the challenge with RMg presented as a hypothesis-free test not restricting itself to a predefined list of pathogens, with utility for the unexpected, the un-requested, the unknown or novel agent [6,7]. These are defining features of infectious diseases investigation, and the source of uncertainty that CMg is uniquely positioned to address. Yet this attribute may prove the most challenging for accrediting and regulatory bodies. Their standards are becoming more demanding with ISO15189:2012, soon to be updated, new accreditation requirements for machine learning components of a diagnostic test with SO14971:2022, and regulatory approvals in some jurisdictions in a transition period moving from the In Vitro Diagnostic Device (IVDD)-Directive to more demanding IVDD-Regulations. Their early and iterative involvement in defining an achievable framework for gathering required evidence is preferable.

Laboratory operational and healthcare system strategic requirements

Most pathogen sequencing is currently performed in academic or public health laboratories but will need to be performed in diagnostic laboratories for routine service. Indeed, there may be advantages moving the dominant sequence capability into the clinical laboratory [60], with data rather than occasional samples transferred to academic and public health laboratories for further analysis (Fig. 2). It would increase the speed and volume of samples sequenced, reduce laborious send-away processes and duplication, and place the first sequence evaluation with staff linked with the presenting case, to optimise co-assessment of pathogen genomics with detailed clinical phenotypes.

Establishing pathogen sequencing within the microbiology laboratory has implications for the target operating model, which in many countries is for consolidation including to off-site facilities, utilising increased automation of culture-based core laboratory workflows. This is anticipated to enhance investment in molecular diagnostics and innovation, while improving quality and turnaround times [61]; however ideally not at the expense of further separating the infection discipline into patient-facing and laboratory-facing components. Pathogen sequencing and metagenomics could unify these requirements, providing comprehensive rapid results in a clinically relevant timeframe, while integrating data output from across a healthcare network for AMR and infection control surveillance benefits.

Every laboratory network will have different factors to consider in placing sequencing capability alongside relevant clinical and scientific expertise, ensuring efficient sample transport, clinical reporting and data infrastructure. For some it may be considered a disruptive additional change overlaid on existing plans. For all, it will require investment that is probably beyond the diagnostic laboratory budget or strategic remit and will need leadership from the healthcare system embedding infectious diseases within its wider genomic, artificial intelligence, and digital strategy [62,63]. It certainly shares with cancer and human genetics the transformative opportunity of personalised medicine alongside the infrastructure, ethical and information governance requirements analysing, storing and transferring genetic data particularly across organisational boundaries. Paradoxically, countries with a less mature existing microbiology infrastructure, may be able to leap-frog the current laboratory model, moving pathogen sequencing faster and more efficiently into their future healthcare system, unrestrained from compromising around the traditional operating model [64].

CONCLUSION

This review summarises the dramatic progress in pathogen sequencing during the COVID-19 pandemic, which served as a reminder that serious emerging and re-emerging infections are frequent events, exacerbated by global warming, biodiversity loss, international trade and travel [64]. When combined with increasing AMR and diversity of opportunistic pathogens in the context of immunosuppression, the need to modernise microbiology services by routine provision of pathogen sequencing is recognised as an even more urgent priority [65]. RMg is moving to the forefront of the pathogen sequencing agenda, based on its ability to link routine testing for personalised patient antimicrobial treatment with improvements in infection control, AMR and public health surveillance and ultimately the societal response to new pandemic threats. Further technology development and laboratory assessment is certainly required transitioning from early clinical evaluation to routine diagnostic provision, but the focus now is more on gathering evidence that the clinical and wider healthcare opportunities of RMg and other pathogen sequencing tests can be cost-effectively realised upon implementation into healthcare systems. Perhaps more importantly, given the transformational change required, is the need for healthcare systems to begin integrating pathogen sequencing alongside cancer genetics and human genomics as part of their wider transformation strategy.

Acknowledgements

None.

Financial support and sponsorship

JE received funding from the National Institute for Health Research (NIHR) Biomedical Research Centre based at Guy's and STtThomas’ National Health Service (NHS) Foundation Trust and Kings College London, the programme of Infection and Immunity (RJ112/N027) and the Guy's and St Thomas’ Fund grant TCF 190910. JE receives research funding from Oxford Nanopore Technologies under a collaboration agreement with Guy's & St Thomas’ Hospital.

Conflicts of interest

JE is employed part-time by Oxford Nanopore Technologies

REFERENCES AND RECOMMENDED READING

Papers of particular interest, published within the annual period of review, have been highlighted as:

▪ of special interest

▪▪ of outstanding interest

REFERENCES 1. Bermingham A, Chand MA, Brown CS, et al. Severe respiratory illness caused by a novel coronavirus, in a patient transferred to the United Kingdom from the Middle East, September 2012. Euro Surveill 2012; 17:1–5. 2. Cho SY, Kang J, Ha YE, et al. MERS-CoV outbreak following a single patient exposure in an emergency room in South Korea: an epidemiological outbreak study. Lancet 2016; 388:994–1001. 3. Zhu N, Zhang D, Wang W, et al. A novel coronavirus from patients with pneumonia in China, 2019. N Engl J Med 2020; 382:727–733. 4. Torres A, Lee N, Cilloniz C, et al. Laboratory diagnosis of pneumonia in the molecular age. Eur Respir J 2016; 48:1764–1778. 5▪. Pandolfo AM, Horne R, Jani Y, et al. Understanding decisions about antibiotic prescribing in ICU: an application of the Necessity Concerns Framework. BMJ Qual Saf 2022; 31:199–210. 6. Govender KN. Precision pandemic preparedness: improving diagnostics with metagenomics. J Clin Microbiol 2021; 59:e02146–e2220. 7. Ko KKK, Chng KR, Nagarajan N. Metagenomics-enabled microbial surveillance. Nat Microbiol 2022; 7:486–496. 8. Simner PJ, Miller S, Carroll KC. Understanding the promises and hurdles of metagenomic next-generation sequencing as a diagnostic tool for infectious diseases. Clin Infect Dis 2018; 66:778–788. 9. Chiu CY, Miller SA. Clinical metagenomics. Nat Rev Genet 2019; 20:341–355. 10. Edgeworth JD, Batra R, Nebbia G, et al. Translational research in the time of COVID-19-dissolving boundaries. PLoS Pathog 2020; 16:e1008898. 11▪. Govender KN, Street TL, Sanderson ND, Eyre DW. Metagenomic sequencing as a pathogen-agnostic clinical diagnostic tool for infectious diseases: a systematic review and meta-analysis of diagnostic test accuracy studies. J Clin Microbiol 2021; 59:e0291620–e291720. 12▪. Liu J, Zhang Q, Dong Y, et al. Diagnostic accuracy of metagenomic next-generation sequencing in diagnosing infectious diseases: a meta-analysis. Sci Rep 2022; 12:1–10. 13▪. Serpa PH, Deng X, Abdelghany M, et al. Metagenomic prediction of antimicrobial resistance in critically ill patients with lower respiratory tract infections. Genome Med 2022; 14:74–84. 14. Anahtar MN, Yang JH, Kanjilal S. Applications of machine learning to the problem of antimicrobial resistance: an emerging model for translational research. J Clin Microbiol 2021; 59:e0126020-20. 15. Schuele L, Cassidy H, Peker N, et al. Future potential of metagenomics in microbiology laboratories. Expert Rev Mol Diagn 2021; 21:1273–1285. 16. Charalampous T, Kay GL, Richardson H, et al. Nanopore metagenomics enables rapid clinical diagnosis of bacterial lower respiratory infection. Nat Biotechnol 2019; 37:783–792. 17. Charalampous T, Alcolea-Medina A, Snell LB, et al. Evaluating the potential for respiratory metagenomics to improve treatment of secondary infection and detection of nosocomial transmission on expanded COVID-19 intensive care units. Genome Med 2021; 13:1–16. 18▪. Baldan R, Cliff PR, Burns S, et al. Development and evaluation of a nanopore 16S rRNA gene sequencing service for same day targeted treatment of bacterial respiratory infection in the intensive care unit. J Infect 2021; 83:167–174. 19. Chapman R, D’Angelo A, Bagby S. Nanopore-based metagenomic sequencing: a diagnostic tool in respiratory tract infection. ERJ Open Res 2022; 8:00461–2022. 20. Govender KN, Eyre DW. Benchmarking taxonomic classifiers with Illumina and Nanopore sequence data for clinical metagenomic diagnostic applications. Microb Genom 2022; 8:1–13. 21. Zhang J, Gao L, Zhu C, et al. Clinical value of metagenomic next-generation sequencing by Illumina and Nanopore for the detection of pathogens in bronchoalveolar lavage fluid in suspected community-acquired pneumonia patients. Front Cell Infect Microbiol 2022; 12:1–11. 22. Chen X, Cao K, Wei Y, et al. Metagenomic next-generation sequencing in the diagnosis of severe pneumonias caused by Chlamydia psittaci. Infection 2020; 48:535–542. 23. Yang X, Wu W, Wang Y, et al. A case of secondary pulmonary syphilis - the utility of mNGS in bronchoalveolar lavage fluid: a case report. Infect Drug Resist 2022; 15:5215–5219. 24. Li T, Chen Y, Lin J, et al. Successful treatment of disseminated nocardiosis diagnosed by metagenomic next-generation sequencing: a case report and review of literature. World J Clin Cases 2022; 10:10120–10129. 25. Liu Y, Ma X, Chen J, et al. Nontuberculous mycobacteria by metagenomic next-generation sequencing: three cases reports and literature review. Front Public Health 2022; 10:1–8. 26. Song P, Li X. Diagnostic value of metagenomics next-generation sequencing technology in disseminated strongyloidiasis. World J Clin Cases 2022; 10:12455–12457. 27. Zhao Y, Ding Y, Zhao X, et al. Role and clinical application of metagenomic next-generation sequencing in immunocompromised patients with acute respiratory failure during veno-venous extracorporeal membrane oxygenation. Front Cell Infect Microbiol 2022; 12:1–7. 28. Zhang A, Xia X, Yuan X, et al. Clinical characteristics of 14 cases of severe Chlamydia psittaci pneumonia diagnosed by metagenomic next-generation sequencing: a case series. Medicine (Baltimore) 2022; 101:e29238. 29. Chen Y, Feng W, Ye K, et al. Application of metagenomic next-generation sequencing in the diagnosis of pulmonary infectious pathogens from bronchoalveolar lavage samples. Front Cell Infect Microbiol 2021; 11:1–10. 30. Mu S, Hu L, Zhang Y, et al. Prospective evaluation of a rapid clinical metagenomics test for bacterial pneumonia. front cell infect microbiol 2021; 11:1–13. 31. Tao Y, Yan H, Liu Y, et al. Diagnostic performance of metagenomic next-generation sequencing in pediatric patients: a retrospective study in a large children's medical center. Clin Chem 2022; 68:1031–1041. 32▪. Chen Y, Fan L, Chai Y, Xu J. Advantages and challenges of metagenomic sequencing for the diagnosis of pulmonary infectious diseases. Clin Respir J 2022; 16:646–656. 33. Diao Z, Han D, Zhang R, Li J. Metagenomics next-generation sequencing tests take the stage in the diagnosis of lower respiratory tract infections. J Adv Res 2021; 38:201–212. 34. Xie F, Duan Z, Zeng W, et al. Clinical metagenomics assessments improve diagnosis and outcomes in community-acquired pneumonia. BMC Infect Dis 2021; 21:352–361. 35▪. Fan S, Si M, Xu N, et al. Metagenomic next-generation sequencing-guided antimicrobial treatment versus conventional antimicrobial treatment in early severe community-acquired pneumonia among immunocompromised patients (MATESHIP): a study protocol. Front Microbiol 2022; 13:927842. 36▪. Tsitsiklis A, Osborne CM, Kamm J, et al. Lower respiratory tract infections in children requiring mechanical ventilation: a multicentre prospective surveillance study incorporating airway metagenomics. Lancet Microbe 2022; 3:e284–e293. 37. Sun H, Wang F, Zhang M, et al. Diagnostic value of bronchoalveolar lavage fluid metagenomic next-generation sequencing in pneumocystis jirovecii pneumonia in non-HIV immunosuppressed patients. Front Cell Infect Microbiol 2022; 12:1–10. 38. Lu X, Zhang J, Ma W, et al. Pneumocystis Jirovecii pneumonia diagnosis via metagenomic next-generation sequencing. Front Med (Lausanne) 2022; 9:1–8. 39. Guo Y, Li L, Li Z, et al. Tropheryma whipplei Detection by nanopore sequencing in patients with interstitial lung disease. Front Microbiol 2021; 12:1–6. 40. Langelier C, Kalantar KL, Moazed F, et al. Integrating host response and unbiased microbe detection for lower respiratory tract infection diagnosis in critically ill adults. Proc Natl Acad Sci U S A 2018; 115:E12353–E12362. 41. Gaston DC, Miller HB, Fissel JA, et al. Evaluation of metagenomic and targeted next-generation sequencing workflows for detection of respiratory pathogens from bronchoalveolar lavage fluid specimens. J Clin Microbiol 2022; 60:e0052622-22. 42. Cocco P, Ayaz-Shah A, Messenger MP, et al. Target Product Profiles for medical tests: a systematic review of current methods. BMC Med 2020; 18:119–121. 43. He P, Wang J, Ke R, et al. Comparison of metagenomic next-generation sequencing using cell-free DNA and whole-cell DNA for the diagnoses of pulmonary infections. Front Cell Infect Microbiol 2022; 12:1–11. 44. Votintseva AA, Bradley P, Pankhurst L, et al. Same-day diagnostic and surveillance data for tuberculosis via whole-genome sequencing of direct respiratory samples. J Clin Microbiol 2017; 55:1285–1298. 45. Ma W, Zhao Y, Lu X, et al. Negative results of bronchoalveolar lavage fluid metagenomic next-generation sequencing in critically ill patients. Front Cell Infect Microbiol 2022; 12:1–9. 46. Public Health England. UK Standards for Microbiology Investigations B 37: investigation of blood cultures (for organisms other than Mycobacterium species). 2022; Available at: https://www.gov.uk/government/publications/smi-b-37-investigation-of-blood-cultures-for-organisms-other-than-mycobacterium-species. [Accessed 10 January 2023]. 47. Public Health England. UK Standards for Microbiology Investigations S 2: pneumonia. 2019; Available at: https://www.gov.uk/government/publications/smi-s-2-pneumonia. [Accessed 10 January 2023]. 48. Torres A, Niederman MS, Chastre J, et al. International ERS/ESICM/ESCMID/ALAT guidelines for the management of hospital-acquired pneumonia and ventilator-associated pneumonia: Guidelines for the management of hospital-acquired pneumonia (HAP)/ventilator-associated pneumonia (VAP) of the European Respiratory Society (ERS), European Society of Intensive Care Medicine (ESICM), European Society of Clinical Microbiology and Infectious Diseases (ESCMID) and Asociación Latinoamericana del Tórax (ALAT). Eur Respir J 2017; 50:1–26. 49. Kalil AC, Metersky ML, Klompas M, et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 clinical practice guidelines by the infectious diseases society of America and the American thoracic society. Clin Infect Dis 2016; 63:e61–e111. 50. Metlay JP, Waterer GW, Long AC, et al. Diagnosis and treatment of adults with community-acquired pneumonia. An official clinical practice guideline of the American thoracic society and infectious diseases society of America. Am J Respir Crit Care Med 2019; 200:e45–e67. 51. Evans L, Rhodes A, Alhazzani W, et al. Surviving sepsis campaign: international guidelines for management of sepsis and septic shock 2021. Crit Care Med 2021; 49:e1063–e1143. 52. Tasbakan MS, Gurgun A, Basoglu OK, et al. Comparison of bronchoalveolar lavage and mini-bronchoalveolar lavage in the diagnosis of pneumonia in immunocompromised patients. Respiration 2011; 81:229–235. 53. Roberts KL, Micek ST, Juang P, Kollef MH. Controversies and advances in the management of ventilator associated pneumonia. Expert Rev Respir Med 2017; 11:875–884. 54. Grgurich PE, Hudcova J, Lei Y, et al. Diagnosis of ventilator-associated pneumonia: controversies and working toward a gold standard. Curr Opin Infect Dis 2013; 26:140–150. 55. Brendish NJ, Malachira AK, Beard KR, et al. Hospitalised adults with pneumonia are frequently misclassified as another diagnosis. Respir Med 2019; 150:81–84. 56. Pettigrew MM, Kwon J, Gent JF, et al. Comparison of the respiratory resistomes and microbiota in children receiving short versus standard course treatment for community-acquired pneumonia. mBio 2022; 13:e0019522-22. 57. Pandolfo AM, Horne R, Jani Y, et al. Intensivists’ beliefs about rapid multiplex molecular diagnostic testing and its potential role in improving prescribing decisions and antimicrobial stewardship: a qualitative study. Antimicrob Resist Infect Control 2021; 10:1–8. 58. Xi Y, Zhou J, Lin Z, et al. Patients with infectious diseases undergoing mechanical ventilation in the intensive care unit have better prognosis after receiving metagenomic next-generation sequencing assay. Int J Infect Dis 2022; 122:959–969. 59. Peabody J, Tran M, Paculdo D, et al. Establishing clinical utility for diagnostic tests using a randomized controlled, virtual patient trial design. Diagnostics (Basel) 2019; 9:1–11. 60. Snell LB, Cliff PR, Charalampous T, et al. Rapid genome sequencing in hospitals to identify potential vaccine-escape SARS-CoV-2 variants. Lancet Infect Dis 2021; 21:1351–1352. 61. Vandenberg O, Durand G, Hallin M, et al. Consolidation of clinical microbiology laboratories and introduction of transformative technologies. Clin Microbiol Rev 2020; 33:e00057–19. 62. Bombard Y, Ginsburg GS, Sturm AC, et al. Digital health-enabled genomics: opportunities and challenges. Am J Hum Genet 2022; 109:1190–1198. 63. Topol Eric. Preparing the healthcare workforce to deliver the digital future. 2019; Available at: https://topol.hee.nhs.uk/wp-content/uploads/HEE-Topol-Review-2019.pdf. [Accessed 10 January 2023]. 64. Gardy JL, Loman NJ. Towards a genomics-informed, real-time, global pathogen surveillance system. Nat Rev Genet 2018; 19:9–20. 65. Topol Eric. A culture of [blood] cultures: Why hasn’t rapid sequencing for serious infections and sepsis become standard of care? 2022; Available at: https://erictopol.substack.com/p/a-culture-of-blood-cultures. [Accessed 10 January 2023].

留言 (0)

沒有登入
gif