Development and implementation of a customised rapid syndromic diagnostic test for severe pneumonia =================================================================================================== * Vilas Navapurkar * Josefin Bartholdson Scott * Mailis Maes * Thomas P Hellyer * Ellen Higginson * Sally Forrest * Joana Pereira-Dias * Surendra Parmar * Emma Heasman-Hunt * Petra Polgarova * Jo Brown * Lissamma Titti * William PW Smith * Jonathan Scott * Anthony Rostron * Matthew Routledge * David Sapsford * M.Estée Török * Ronan McMullan * David A Enoch * Vanessa Wong * The VAPrapid investigators * Martin D Curran * Nicholas M Brown * A John Simpson * Jurgen Herre * Gordon Dougan * Andrew Conway Morris ## Abstract **Background** Microbial cultures for the diagnosis of pneumonia take several days to return a result, and are frequently negative, compromising antimicrobial stewardship. The objective of this study was to establish the performance of a syndromic molecular diagnostic approach, using a custom TaqMan array card (TAC) covering 52 respiratory pathogens, and assess its impact on antimicrobial prescribing. **Methods** The TAC was validated against a retrospective multi-centre cohort of broncho-alveolar lavage samples. The TAC was assessed prospectively in patients undergoing investigation for suspected pneumonia, with a comparator cohort formed of patients investigated when the TAC laboratory team were unavailable. Co-primary outcomes were sensitivity compared to conventional microbiology and, for the prospective study, time to result. Metagenomic sequencing was performed to validate findings in prospective samples. Antibiotic free days (AFD) were compared between the study cohort and comparator group. **Results** 128 stored samples were tested, with sensitivity of 97% (95% CI 88-100%). Prospectively 95 patients were tested by TAC, with 71 forming the comparator group. TAC returned results 51 hours (IQR 41-69 hours) faster than culture and with sensitivity of 92% (95% CI 83-98%) compared to conventional microbiology. 94% of organisms identified by sequencing were detected by TAC. There was a significant difference in the distribution of AFDs with more AFDs in the TAC group (p=0.02). TAC group were more likely to experience antimicrobial de-escalation (OR 2.9 (95%1.5-5.5). **Conclusions** Implementation of a syndromic molecular diagnostic approach to pneumonia led to faster results, with high sensitivity and impact on antibiotic prescribing. **Trial registration** The prospective study was registered with [clinicaltrials.gov](http://clinicaltrials.gov) [NCT03996330](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT03996330&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F11%2F2020.06.02.20118489.atom) Keywords * Antimicrobial stewardship * Critical Care * Molecular pathology * Pneumonia ## Introduction For many decades the diagnosis of infectious diseases has relied on a combination of clinical assessment and microbiological culture. However, cultures are frequently negative 1,2 and can take several days to return a result.3 Optimising antimicrobial therapy can be challenging, especially in patients who are at risk of multidrug resistant organisms.2 In critically ill patients, this frequently results in the empiric use of broad-spectrum agents, with predictable consequences for antimicrobial resistance and other forms of antimicrobial-related harm.4 Conversely, failure to identify the causative organism can lead to inappropriate antimicrobial therapy, which is associated with poor outcomes.5 Pneumonia amongst intubated and mechanically ventilated, critically ill patients can be especially difficult to diagnose.6 Most critically ill patients are systemically inflamed,7 clinical examination is unreliable8 and there are multiple causes of radiographic lung infiltrates most of which are non-infectious.9,10 The development of host-based biomarkers for infection, such as C-reactive protein,11 procalcitonin,12 and alveolar cytokine concentrations13 have been advanced as useful measures to help rationalise antimicrobial use. However, their utility in the diagnosis11,12 and antimicrobial stewardship14,15 of pneumonia has been challenged. There is, therefore, a pressing need for rapid, sensitive, multi-pathogen-focussed diagnostic tests for pneumonia16. TaqMan array cards (TAC) enable the conduct of multiple simultaneous single-plex real-time polymerase chain reaction (RT-PCR), with this format allowing rapid and straightforward customisation. Although TACs have shown promising performance relative to conventional microbiology,17 our previous experience demonstrated that a TAC with restricted coverage of common respiratory pathogens had a limited impact on clinical decision making in critically ill patients.18 We therefore set out to develop and implement a multi-pathogen array that would have broad applicability for severe pneumonia. ## Materials and Methods ### Card development Organism selection was informed by review the literature concerning organisms found in severe pneumonia 1,2,6,13,18,19. Details of organism selection and the card layout are shown in supplemental section (Supplemental Figure S1). The card covers 52 organisms (23 bacteria, 2 mycobacteria, 6 atypical bacteria, 5 fungi and 16 viruses). The study was undertaken prior to the COVID-19 pandemic starting in 2020. ### Card validation #### Technical validation The card was initially validated against stored extracts, synthetic control plasmids and all available External Quality Assessment (EQA) panels from Quality Control for Molecular Diagnostics ([www.qcmd.org](http://www.qcmd.org)) (supplemental results). ### Retrospective cohort validation A retrospective cohort validation was conducting using stored bronchoalveolar lavage (BAL) samples obtained during the twenty four centre VAPrapid trial of a biomarker for the diagnosis of ventilator-associated pneumonia.15 VAPrapid centres used semi-quantitative microbiological culture as the reference standard. ### Prospective evaluation #### Setting Patients were recruited from a 20-bedded teaching hospital Intensive Care Unit (ICU). The unit is a mixed general medical-surgical unit which supports organ and haematology-oncology services. ### Recruitment Between February 2018 and August 2019, prospectively identified patients were eligible for inclusion if they were receiving invasive mechanical ventilation, and if the treating intensive care specialist was performing diagnostic bronchoscopy for suspected pneumonia. Written consent was obtained from the patient or a proxy decision maker. The TAC laboratory team were routinely unavailable from Friday 5pm to Monday 8am, and also sporadically unavailable due to leave. Patients who were not included in the study because of a lack of TAC laboratory team availability, and those from the month prior and month following the study, formed the comparator group. ### Sampling procedure Bronchoscopy was undertaken in accordance with existing unit protocols (supplemental methods). ### TAC testing Nucleic acids were extracted from BAL prior to loading on the TAC. The TAC was run by a dedicated laboratory team who did not undertake the conventional PCR or cultures, with blinding also assured by the results of the TAC being obtained before those from conventional microbiology. Full details of the TAC process are included in the supplemental section. ### Conventional microbiological testing BAL samples were processed according to the UK Standards for Microbiology Investigations (SMI),20 with the results of microbiological semi-quantitative culture and conventional PCR for respiratory viruses, herpesvirade and *Pneumocystis jirovecii* forming the reference standard for the TAC (supplemental methods). As an experimental assay, the results of the TAC were not included in the laboratory information system, blinding the assessors of the reference standard to the TAC results. ### Return of results to clinical team Following review by a consultant clinical scientist, results were returned to the ICU team. Clinical microbiology advice was available 24 hours/day, and patients underwent weekday daily combined ICU-Microbiology multi-disciplinary review in keeping with existing unit practice (weekend microbiology input was available on request). The study did not mandate any course of action by the treating clinical team. Conventional microbiology results were returned to clinicians via the electronic health record; however in practice these were returned after the TAC results. ### Outcome measures The co-primary outcome measures were sensitivity, using conventional microbiology as the reference standard and time to result compared to conventional microbial culture. Time to result for microbial culture was taken as time from completion of lavage to first organism identification, or confirmation of negative growth if no organisms were detected. Secondary outcome measures were sensitivity compared to metagenomic microbial sequencing, time to result compared to conventional PCR, days alive and free of antibiotics (antibiotic-free days, AFDs) in seven and twenty-eight days following lavage and change in antibiotic therapy in the seven days following lavage. Qualitative assessment of whether TAC results impacted on antimicrobial change was assessed by clinical notes review by a member of the study team who was not involved in the decision-making process (VW). ### Statistical analysis The difference in median time to result for conventional culture and TAC was assessed by Wilcoxon’s matched-pairs test. Where conventional PCR failed, or where the lab did not test for the organism, the corresponding tests from the TAC were removed from calculation of diagnostic performance. Indeterminate cultures (‘mixed upper respiratory tract flora’) were considered negative. A sensitivity analysis, coding failed conventional PCR and organisms not tested ‘negative’ was also undertaken. Comparisons of distribution of antibiotic free days between TAC and comparator groups was by Mann-Whitney U test, differences in proportions of escalation and de-escalation decisions were assessed by Chi2 test. Analyses were conducted using Prism v9.1 (Graphpad Inc, La Jolla, CA). ### Study size A planned prospective study size of 100 patients evaluated by TAC was selected to balance cost against including sufficient numbers to be able to make a judgement on the card’s clinical utility. As the co-primary endpoint was time to result in a real-world setting that had not been previously evaluated, we did not undertake a formal power calculation. ### Ethical and regulatory approvals and funding The prospective study was approved by the Leeds East Research Ethics Committee (17/YH/0286) Cambridge University Hospitals NHS Foundation Trust was the sponsor, and registered with [clinicaltrials.gov](http://clinicaltrials.gov) ([NCT03996330](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT03996330&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F11%2F2020.06.02.20118489.atom)). The assessment of routinely collected data from the comparator group received a consent waiver and was conducted under a protocol approved by the institutional review board (A095506). VAPrapid15 was approved by the England and Northern Ireland (13/LO/065) and Scotland (13/SS/0074) National Research Ethics Service committees and sponsored by Newcastle upon Tyne Hospitals NHS Foundation Trust. ## Results ### Technical validation Following initial validation against stored DNA extracts and synthetic plasmids, all microorganisms from the Quality Control for Molecular Diagnostics 2018 Sepsis EQA Pilot Study were successfully detected (supplemental Table S1). ### Retrospective cohort validation The card was tested against the stored samples available from the VAPrapid study15. 128 samples with semi-quantitative culture results were available for analysis. 57 organisms were grown at or above 104 colony forming units(CFU)/ml 20,21, with 55 detected by TAC (Table 1). The TAC detected a further 295 organisms, including 64 viruses and one atypical organism which the recruiting centres did not test for. Excluding tests for organisms not detectable by culture, 3425 tests on TAC were negative. Sensitivity was 97% (95% CI 88-100%) and specificity 94% (95% CI 93-95%) (Supplemental Table S2). Organisms detected by both TAC and culture had a median cycles to threshold (Ct) value on the TAC of 29 (IQR 26-32 range 20-35) whilst those detected by TAC alone had a median Ct value of 33 (IQR 30-35 range 20-40) (supplemental Figure S2). View this table: [Table 1:](http://medrxiv.org/content/early/2021/06/11/2020.06.02.20118489/T1) Table 1: culture of microorganisms from 128 stored samples from the VAPrapid clinical trial15 and results from the TAC. CFU-colony forming units/ml, Ct -cycles to crossing threshold. *not on card # culture reported as *Proteus mirabilus*, on TAC reported as genus-level *Proteus spp.* ### Prospective evaluation Between January 2018 and September 2019 166 ventilated patients were investigated for pneumonia by bronchoscopy, 95 were tested by TAC. Five patients were tested twice by TAC, having suffered a subsequent respiratory deterioration, in total 100 TACs were run. 71 patients formed the comparator group (Figure 1). Although inclusion criteria were pragmatic and only required senior clinician suspicion of pneumonia, 92% of cases met full ECDC criteria for clinical pneumonia (Supplemental Figure S3). Of the eight cases not meeting full ECDC criteria, one lacked a formal radiological report of infiltrates, one had no clinical signs of pneumonia, five had no signs of systemic inflammation and one patient lacked both radiological and systemic inflammation. Table 2 shows participant characteristics of the study population and comparator group. View this table: [Table 2:](http://medrxiv.org/content/early/2021/06/11/2020.06.02.20118489/T2) Table 2: Baseline characteristics of study population. APACHE II, acute physiology and chronic health evaluation II, FiO2, fraction of inspired oxygen. ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/06/11/2020.06.02.20118489/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2021/06/11/2020.06.02.20118489/F1) Figure 1: Study flow diagram *included in comparator group as TAC laboratory team not available to process samples ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/06/11/2020.06.02.20118489/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2021/06/11/2020.06.02.20118489/F2) Figure 2: Distribution of days alive and free of antibiotics in the seven days following bronchoscopy and lavage in the TAC and comparator cohorts. Following first lavage only for patients who had more than one BAL during ICU admission. Numbers in each category and percentage shown below graph, p value by Mann-Whitney U test. ### Time to result The median difference in time to result between TAC and conventional culture was 51 hours (IQR 41-69 hours p<0.0001 by Wilcoxon matched pairs), the TAC also returned results more rapidly than conventional PCR in almost all cases (Supplemental Figure S4). The minimum TAC time to return was 4 hours, with median time to result 22 hours (IQR 7-24 hours), most of the delays arose from samples taken outside routine working hours, whilst additional delays with conventional PCR results largely reflect laboratory workflow and batching of samples. ### Comparison of organisms detected by TAC compared to conventional microbiology 178 organisms were identified from 100 samples on the TAC (Table 3, Supplemental Table S3). Conventional microbiology detected 66 organisms, with 61 detected by TAC. 27 patients had failure of internal control for one or more conventional PCR assays, covering 93 organisms. There were no TAC internal control failures and none of the organisms covered by the failed assays were detected on TAC or sequencing (Table 3). Sensitivity and specificity were 92% (95% CI 83-98%) and 97% (95% CI 97-98%) respectively (Supplemental Table S4). Including failed and absent reference standards as ‘negative’ had minimal effect on diagnostic performance (Supplemental Table S5) View this table: [Table 3:](http://medrxiv.org/content/early/2021/06/11/2020.06.02.20118489/T3) Table 3: Summary of organisms detected by conventional microbiological testing (left hand column), by TAC (middle column), and by microbial sequencing (right hand column). * One hit not found in same patient; not on card. **Legionella urinary antigen test positive. \***| Positive BAL galactomannan enzyme immunoassay (>0.5 units) with CT consistent with fungal pneumonia and known risk factors but fungal cultures were not positive. # refers to human coronavirade OC43, 229E and NL63, no tests were undertaken for SARS-CoV2 and final testing occurred in August 2019. ### Comparison by sequencing 98 samples were available for sequencing. Metagenomic sequencing revealed 107 organisms, 100 of which were also detected by TAC (Tables 3, S3). Concerning the 10 organisms detected by conventional microbiology or sequencing but missed by TAC, one organism, that was positive by both culture and sequencing albeit in different patients, was *Citrobacter freundii*, for which we did not have a sequence on the card. A further five pathogens were detected by sequencing (*Staphylococcus aureus, Legionella* spp., and *Staphylococcus epidermidis)* or both culture and sequencing (two *E. faecium*). Although these five were detected by TAC, they did not pass the internal quality control standards required for reporting and were considered ‘negative’ results. The remaining three organisms, two rhinovirus by conventional PCR and one *Staphylococcus* spp. by sequencing, were not detected by TAC at all. One case of *Aspergillus fumigatus* was detected on the TAC, and although no moulds were cultured, the lavage galactomannan antigen test was highly positive (5.92 optical density index (ODI), laboratory reference range <0.5 ODI). ### Quantitation Twenty-five organisms were grown on conventional culture at ≥104 CFU/ml, the conventional cut off for quantitative culture of lavage.20, 21 The median cycles to threshold (Ct) for these organisms on the TAC was 27 (IQR 24-29, range 20-33). In contrast, culturable organisms detected on TAC but not on culture had a median Ct of 32 (IQR 30-34, range 22-38) (supplemental Figure S2). ### Antibiotic prescribing Patients in the TAC and comparator cohorts had similar severity of illness, severity of respiratory failure and demographic features (Table 2). Patients managed with the TAC had a significantly different distribution of AFDs to the comparator group in the 7 days following bronchoscopy (p=0.02 by Mann-Whitney U-test), with more AFDs in the TAC cohort. This difference did not retain significance over 28 days (Supplemental Figure S5). Overall 72 (76%) of TAC patients had their antibiotics changed in the seven days following bronchoscopy, with a total of 116 changes made (Table 4). In the comparator group 50 (70%) of patients experienced a total of 65 changes. Whilst 63% of decisions in the TAC group led to de-escalation, only 37% of decisions in the comparator group were de-escalation decisions (OR 2.9 (95% CI 1.5-5.5) p=0.008 by Chi-squared). Decisions which were judged to be related to the TAC result were weighted further towards de-escalation (73% of all TAC-related changes, Table 4). 11 (30%) of escalations in the TAC group were judged to have been targeted escalations in response to TAC results. In a further six cases negative TAC results prompted investigation for alternative diagnoses. View this table: [Table 4:](http://medrxiv.org/content/early/2021/06/11/2020.06.02.20118489/T4) Table 4: Detail of changes in antibiotic therapy in the seven days following lavage in the TAC and comparator cohorts. Changes judged to be TAC-related are shown in the left-hand sub-column for the TAC group. Several patients had more than one change in antibiotic therapy. *includes two de-escalations to prophylactic dose, ** includes two escalations from prophylactic to therapeutic dose. ## Discussion We demonstrate that a customised molecular diagnostic, designed to meet the needs of a specific clinical setting produced accurate results in a clinically important time-frame and was associated with an increase in antibiotic-free days relative to the comparator group in the week following investigation. Diagnostic performance was similar when assessed in stored samples from multiple centres, indicating a generalisable result. Molecular diagnostic platforms for respiratory infection syndromes have, until recently, largely focussed on viral pathogens.16 However, the need to optimise antimicrobial therapy whilst limiting the over-use of these drugs has led to repeated calls for bacterial-focussed diagnostics.16,22 TACs have been previously reported for use in pneumonia.18,23, 24 However, apart from our previous report18 that demonstrated limited clinical impact due to restricted organism coverage, none of the other reports have included ventilated patients and were restricted to retrospective analysis of stored samples. Commercial multiple-pathogen arrays that include respiratory bacteria have recently become available. However, most of reports of their use in ventilated patients remain limited to describing diagnostic performance, reporting ‘potential’ to change antimicrobial therapy rather than impact on clinical practice3,25,26. Concerns have been raised about the risks of over-treatment from molecular diagnostics16,27,28, whilst conversely promising tests with the potential to change therapy have not always proven this in clinical practice15,18. These commercially available assays lack the broad coverage and customisability of the TAC, with consistent concerns raised around limited organism coverage adversely impacting treatment decisions.3,18,25,26 Although there is now widespread acceptance of the presence of a respiratory microbiome,29,30 the lungs of ventilated patients present a challenge to highly sensitivemolecular diagnostics16,. The proximal respiratory tract of ventilated patients becomes rapidly colonised with predominantly Gram negative organisms.31,32 This can occur in the absence of infection, and there is a risk that highly sensitive techniques will detect colonising organisms, driving unintended increases in antimicrobial use.16 The use of protected lower airway specimens, with growth ≥104 CFU/ml for BAL have been used to distinguish infection from colonisation.21,33 We adapted this approach in this study, using the quasi-quantitative Ct value provided by RT-PCR and testing protected bronchoalveolar samples. Using the comparison of the Ct values of organisms detected by culture and those detected by TAC without culture, we suggest that a Ct threshold of 32 be used to identify infecting rather than colonising organism (supplemental Figures S2, S6). One of the problems that has beset bacterial diagnostics studies has been the absence of a ‘gold standard’ against which the candidate can be assessed,16,22,34 as conventional culture is imperfect. For this study we used a combination of conventional microbiology (culture and viral PCR) and metagenomic sequencing. 10 organisms identified by conventional microbiology or sequencing were not ‘detected by the TAC. Overall the TAC detected more organisms than either culture or sequencing, reflecting the higher sensitivity of qPCR. However, without a perfect validation method we cannot be certain these were not ‘false positives’ and have counted them as such for the calculation of specificity. The sequencing and culture results give clinicians considerable confidence in the results provided. The selection of organisms targeted on the card was crucial, and informed by our previous experience where omission of key organisms significantly limited the impact of a similar card.18 Given the case mix of our unit, with a high proportion of immunosuppressed patients, we opted to include a number of low pathogenicity organisms, (i.e. coagulase-negative *Staphylococci(CNS), Enterococci* and *Candida albicans*), as well as Herpesviridae, which we routinely tested for. The detection of these organisms can be challenging to interpret35, given that many critically ill patients have a degree of immunoparesis, even if not classically immunosuppressed, 6,36 their significance remains uncertain. As our laboratory routinely reported these organisms on conventional microbiology the clinical team were already confronted with this issue. The inclusion of CNS also aids with the interpretation of the detection of the mecA gene, which is commonly carried by these organisms, thus helping identify MRSA. The lack of CNS on commercial cards has been noted to impair interpretation of mecA in clinical samples25,37. However the ready customisability of the TAC would allow units to remove such organisms, as well as add other organisms of local significance as we have done subsequently during the COVID-19 pandemic38. The use of a contemporaneous comparator cohort allowed for comparisons of antibiotic prescribing within the context of the implementation of the TAC and any heightened awareness of antimicrobial stewardship it may have engendered. Despite this, the comparator cohort saw a greater proportion of escalation decisions in the week following lavage, and had fewer antibiotic-free days. The lack of difference in AFDs at day 28 is unsurprising, as suspected pneumonia is only one of multiple drivers of antibiotic use. Although the comparator and TAC groups had similar characteristics, our observational design means that we cannot be certain that unmeasured confounders did not contribute to the effects seen. This study established a molecular diagnostic test to meet the needs of a particular intensive care unit, and implemented it in the context of a well-established antimicrobial stewardship program. Although the demonstration of similar performance on stored samples from multiple centres is reassuring, the impact on antimicrobial stewardship is likely to be context-dependent. Replication in additional settings with distinct approaches to stewardship is required before we can be certain of its external generalisability, whilst evaluation in a randomised, controlled trial would help reduce any bias that may have arisen from our observational study design. We believe this approach represents a promising new approach to the management of severe pneumonia. ## Supporting information Supplemental methods, results, tables and figures [[supplements/118489_file04.docx]](pending:yes) ## Data Availability De-identified data is available on request to the corresponding author. All samples were sequenced at the Wellcome Sanger Institute, and raw read data is available at the European Nucleotide Archive (ENA) with study accession numbers ERP111277, ERP111280, ERP112277, and ERP018622. ## Acknowledgements The VAPrapid investigators are Prof DF McAuley, Prof TS Walsh, Dr N Anderson, Dr S Singh, Prof P Dark, Dr A Roy, Prof GD Perkins, Ms L Emerson, Prof B Blackwood, Dr SE Wright, D K Kefala, Prof CM O’Kane, Dr SV Baudouin, Dr RL Patterson, Dr A Agus, Dr J Bannard-Smith, Dr NM Robbin, Prof ID Welters, Dr C Bassford, Dr B Yates, Dr C Spencer, Dr SK Laha, Dr J Hulme, Prof S Bonner, Dr V Linnett, Dr J Sonsken, Dr Van Den Broeck, Dr G Boschman, Mr DWJ Kennan, Dr AJ Allan, Mr G Phair, Ms J Parker and Dr SA Bowett. The authors thank the consultant intensivists of the John V Farman Intensive Care Unit, Drs P Bradley, P Featherstone, S Ford, M Georgieva, A Johnston, R Mahroof, J Martin, J Preller, K Patel, C Summers, M Trivedi, J Varley, Pharmacist L Radford, the nursing and physiotherapy teams who managed the patients, and the patients and their families who consented to the study. We also thank Torsten Seemann for access to his Kraken database. ## Footnotes * **Declarations Ethical approval and consent to participate**: Written informed consent was obtained from patients or their proxy decision maker for studies where samples were obtained, where participants regained capacity retrospective consent was sought. The samples for the retrospective study were obtained from the VAPrapid study (ref 15) which was approved by the England and Northern Ireland (13/LO/065) and Scotland (13/SS/0074) National Research Ethics Service committees and sponsored by Newcastle upon Tyne Hospitals NHS Foundation Trust. * The prospective study was approved by the Leeds East Research Ethics Committee (17/YH/0286) Cambridge University Hospitals NHS Foundation Trust was the sponsor. The assessment of routinely collected data from the comparator group received a consent waiver and was conducted under a protocol approved by the institutional review board (A095506). * **Consent for publication**: no individual patient data presented, not required * **Availability of data**: Microbial sequencing data is available from European Nucleotide Archive (ENA) with study accession numbers ERP111277, ERP111280, ERP112277, and ERP018622. Patient data is not publicly available for confidentiality reasons, but anonymised data can be obtained through contact with the corresponding author, subject to appropriate data sharing agreements being in place. * **Competing interests**: MDC is the inventor on a patent held by the Secretary of State for Health (UK Government) EP2788503, which covers some of the genetic sequences used in this study. VN is a founder, Director and shareholder in Cambridge Infection Diagnostics Ltd (CID Ltd) which is a commercial company aimed at developing molecular diagnostics in infection and antimicrobial and AMR stewardship. NMB, GD and ACM are members of the Scientific Advisory Board of Cambridge Infection Diagnostics Ltd (CID Ltd). All other authors declare no conflict of interest. * **Funding** The study was funded by Addenbrooke’s Charitable Trust and the NIHR Cambridge Biomedical Resource Centre (Grant 18135 to Professor Dougan). Dr Török is supported by a Clinician Scientist Fellowship (funded by the Academy of Medical Sciences and the Health Foundation) and by the NIHR Biomedical Research Centre. Dr Conway Morris was supported by a Clinical Research Career Development Fellowship from the Wellcome Trust at the time of the work (WT 2055214/Z/16/Z) and is currently supported by an MRC Clinician Scientist Fellowship (MR/V006118/1). The metagenomic sequencing was funded by the Wellcome Trust. VAPrapid was funded Health Innovation Challenge Fund(HICF-510-078;094949/Z/10/X), a parallel funding partnership between the UK Department of Health and Wellcome Trust. Professor Simpson is a National Institute for Health Research (NIHR) Senior Investigator. The views expressed in this publication are those of the authors and not necessarily those of the UK Department of Health or Wellcome Trust. The funders had no role in the analysis of data or decision to publish. The corresponding author had full access to all the data in the study and had final responsibility for the decision to submit for publication. * VN -Conceptualisation, resources, investigation, writing-review and editing, project administration, funding acquisition, supervision. * JBS -resources, investigation, data curation, formal analysis, writing-original draft. * MM-resources, investigation, writing-review and editing. * TH-resources, investigation, writing-review and editing * EH-investigation, writing-review and editing. * SF-investigation, writing-review and editing. * JD-investigation, writing-review and editing. * SP-investigation, writing-review and editing. * EHH-investigation, writing-review and editing. * PP-investigation, data curation writing-review and editing. * JB-investigation, data curation writing-review and editing. * LT-investigation, data curation writing-review and editing. * WS- data curation writing-review and editing * JS- resources, investigation, writing-review and editing * AR- resources, investigation, writing-review and editing * MR-data curation writing-review and editing. * DS-data curation writing-review and editing. * MET-conceptualisation, investigation, data curation writing-review and editing * DE-investigation, writing-review and editing. * VW-formal analysis, investigation, data curation writing-review and editing. * MDC-Conceptualisation, resources, investigation, writing-review and editing, project administration, funding acquisition, supervision. * NB-Conceptualisation, resources, investigation, writing-review and editing, project administration, supervision. AJS- resources, investigation, writing-review and editing * JH-Conceptualisation, resources, investigation, writing-review and editing. * GD-Conceptualisation, resources, investigation, writing-review and editing, project administration, funding acquisition, supervision. * ACM-Conceptualisation, methodology, resources, investigation, writing-original draft, project administration, formal analysis, funding acquisition, supervision. * Additional data added regarding validation of TAC in a cohort of stored samples from the VAPrapid trial (table 1, methods and results, supplemental data), further analysis of antibiotic changes in the prospective and comparator cohorts (table 4 and supplemental section) * Received June 2, 2020. * Revision received June 10, 2021. * Accepted June 11, 2021. * © 2021, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution 4.0 International), CC BY 4.0, as described at [http://creativecommons.org/licenses/by/4.0/](http://creativecommons.org/licenses/by/4.0/) ## References 1. 1.Cilloniz C, Ewig S, Polverino E, et al. Microbial aetiology of community-acquired pneumonia and its relation to severity. Thorax. 2011;66(4):340–346. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToidGhvcmF4am5sIjtzOjU6InJlc2lkIjtzOjg6IjY2LzQvMzQwIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDYvMTEvMjAyMC4wNi4wMi4yMDExODQ4OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 2. 2.Vincent J-L, Sakr Y, Singer M, et al. Prevalence and Outcomes of Infection Among Patients in Intensive Care Units in 2017. JAMA. 2020;323(15):1478–10. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2020.2717&link_type=DOI) 3. 3.Gadsby NJ, McHugh MP, Forbes C, et al. Comparison of Unyvero P55 Pneumonia Cartridge, in-house PCR and culture for the identification of respiratory pathogens and antibiotic resistance in bronchoalveolar lavage fluids in the critical care setting. Eur J Clin Microbiol Infect Dis. 2019;38(6):1171_1178. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s10096-019-03526-x&link_type=DOI) 4. 4.Arulkumaran N, Routledge M, Schlebusch S, Lipman J, Morris AC. Antimicrobial-associated harm in critical care: a narrative review. Intensive Care Med. January 2020:1–11. 5. 5.Iregui M, Ward S, Sherman G, Fraser VJ, Kollef MH. Clinical Importance of Delays in the Initiation of Appropriate Antibiotic Treatment for Ventilator-Associated Pneumonia. Chest 2002;122(1):262–268. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1378/chest.122.1.262&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12114368&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F11%2F2020.06.02.20118489.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000176927900042&link_type=ISI) 6. 6.Morris AC. Management of pneumonia in intensive care. J Emerg Crit Care Med. 2018;2:101–101. 7. 7.Rangel-Frausto MS, Pittet D, Costigan M, Hwang T, Davis CS, Wenzel RP. The Natural History of the Systemic Inflammatory Response Syndrome (SIRS). JAMA. 1995;273(2):117–5. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.1995.03520260039030&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=7799491&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F11%2F2020.06.02.20118489.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1995PZ68700024&link_type=ISI) 8. 8.Lichtenstein D, Goldstein I, Mourgeon E, Cluzel P, Grenier P, Rouby J-J. Comparative diagnostic performances of auscultation, chest radiography, and lung ultrasonography in acute respiratory distress syndrome. Anesthesiology. 2004;100(1):9–15. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/00000542-200401000-00006&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=14695718&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F11%2F2020.06.02.20118489.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000187623200004&link_type=ISI) 9. 9.Meduri GU, Mauldin GL, Wunderink RG, et al. Causes of fever and pulmonary densities in patients with clinical manifestations of ventilator-associated pneumonia. Chest. 1994;106(1):221–235. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1378/chest.106.1.221&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8020275&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F11%2F2020.06.02.20118489.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1994NX37200039&link_type=ISI) 10. 10.Conway Morris A, Kefala K, Wilkinson TS, et al. Diagnostic importance of pulmonary interleukin-1{beta} and interleukin-8 in ventilator-associated pneumonia. Thorax. 2010;65(3):201–207. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToidGhvcmF4am5sIjtzOjU6InJlc2lkIjtzOjg6IjY1LzMvMjAxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDYvMTEvMjAyMC4wNi4wMi4yMDExODQ4OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 11. 11.Povoa P, Coelho L, Almeida E, et al. C-reactive protein as a marker of ventilator-associated pneumonia resolution: a pilot study. Eur Resp J. 2005;25(5):804–812. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiZXJqIjtzOjU6InJlc2lkIjtzOjg6IjI1LzUvODA0IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDYvMTEvMjAyMC4wNi4wMi4yMDExODQ4OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 12. 12.Pfister R, Kochanek M, Leygeber T, et al. Procalcitonin for diagnosis of bacterial pneumonia in critically ill patients during 2009 H1N1 influenza pandemic: a prospective cohort study, systematic review and individual patient data meta-analysis. Critical Care. 2014;18(2):1–11. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13054-014-0528-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F11%2F2020.06.02.20118489.atom) 13. 13.Hellyer TP, Morris AC, Mcauley DF, et al. Diagnostic accuracy of pulmonary host inflammatory mediators in the exclusion of ventilator-acquired pneumonia. Thorax. 2015;70(1):41–47. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToidGhvcmF4am5sIjtzOjU6InJlc2lkIjtzOjc6IjcwLzEvNDEiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8wNi8xMS8yMDIwLjA2LjAyLjIwMTE4NDg5LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 14. 14.Huang DT, Yealy DM, Filbin MR, et al. Procalcitonin-Guided Use of Antibiotics for Lower Respiratory Tract Infection. N Engl J Med. 2018;379(3):236–249. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1802670&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29781385&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F11%2F2020.06.02.20118489.atom) 15. 15.Hellyer TP, McAuley DF, Walsh TS, et al. Biomarker-guided antibiotic stewardship in suspected ventilator-associated pneumonia (VAPrapid2): a randomised controlled trial and process evaluation. Lancet Resp Med. December 2019:1–10. 16. 16.Hanson KE, Azar MM, Banerjee R, et al. Molecular Testing for Acute Respiratory Tract Infections: Clinical and Diagnostic Recommendations From the IDSA’s Diagnostics Committee. Clin Infect Dis. 2020;5:401–408. 17. 17.Steensels D, Reynders M, Descheemaeker P, et al. Clinical evaluation of a multi-parameter customized respiratory TaqMan® array card compared to conventional methods in immunocompromised patients. J Clin Virol. 2015;72:36–41. 18. 18.Jones N, Conway Morris A, Curran MD et al. Evaluating the use of a 22-pathogen TaqMan array card for rapid diagnosis of respiratory pathogens in Intensive Care. J Med Microbiol. 2020; 69: 971–978. 19. 19.Gadsby NJ, Russell CD, McHugh MP, et al. Comprehensive Molecular Testing for Respiratory Pathogens in Community-Acquired Pneumonia. Clin Infect Dis. 2016;62(7):817–823. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/cid/civ1214&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26747825&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F11%2F2020.06.02.20118489.atom) 20. 20.Public Health England. Investigation of bronchoalveolar lavage, sputum and associated specimens UK SMI B 57 issue 3.5 (May 2019) (available from [https://www.gov.uk/government/publications/smi-b-57-investigation-of-bronchoalveolar-lavage-sputum-and-associated-specimens](https://www.gov.uk/government/publications/smi-b-57-investigation-of-bronchoalveolar-lavage-sputum-and-associated-specimens)) accessed 29/4/2021 21. 21.Plachouras D, Lepape A, Suetens C. ECDC definitions and methods for the surveillance of healthcare-associated infections in intensive care units. Intensive Care Med. October 2018:1–3. 22. 22.The Lancet Respiratory Medicine. Pneumonia research: time to fill in the gaps. Lancet Resp Med. 2019;7(12):993. 23. 23.1. Onderdonk AB Wolff BJ, Bramley AM, Thurman KA, et al. Improved Detection of Respiratory Pathogens by Use of High-Quality Sputum with TaqMan Array Card Technology. Onderdonk AB, ed. J Clin Microbiol. 2017;55(1):110–121. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNtIjtzOjU6InJlc2lkIjtzOjg6IjU1LzEvMTEwIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDYvMTEvMjAyMC4wNi4wMi4yMDExODQ4OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 24. 24.1. Schildgen O Hercik C, Cosmas L, Mogeni OD, et al. A diagnostic and epidemiologic investigation of acute febrile illness (AFI) in Kilombero, Tanzania. Schildgen O, ed. PLoS ONE. 2017;12(12):e0189712–e0189720. 25. 25.Peiffer-Smadja N, Bouadma L, Mathy V, et al. Performance and impact of a multiplex PCR in ICU patients with ventilator-associated pneumonia or ventilated hospital-acquired pneumonia. Crit Care. 2020;24:366. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13054-020-03067-2&link_type=DOI) 26. 26.Monard C, Pehlivan J, Auger G, et al. Multicenter evaluation of a syndromic rapid multiplex PCR test for early adaptation of antimicrobial therapy in adult patients with pneumonia. Crit Care. 2020;24:434 27. 27. Houben Rmgj, Lalli M, Kranzer K, Menzies NA, Schumacher SG, Dowdy DW. What if They Don’t Have Tuberculosis? The Consequences and Trade-offs Involved in False-positive Diagnoses of Tuberculosis. Clin Infect Dis. 2019;68:150–156 28. 28.Weinrib DA, Capraro GA. The Uncertain Clinical Benefit of the T2Bacteria Panel. Ann Intern Med. 2019;170:888–889 29. 29.Nolan TJ, Gadsby NJ, Hellyer TP, et al. Low-pathogenicity Mycoplasma spp. alter human monocyte and macrophage function and are highly prevalent among patients with ventilator-acquired pneumonia. Thorax. 2016;71(7):594–600. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToidGhvcmF4am5sIjtzOjU6InJlc2lkIjtzOjg6IjcxLzcvNTk0IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDYvMTEvMjAyMC4wNi4wMi4yMDExODQ4OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 30. 30.Zakharkina T, Martin-Loeches I, Matamoros S, et al. The dynamics of the pulmonary microbiome during mechanical ventilation in the intensive care unit and the association with occurrence of pneumonia. Thorax. 2017;72(9):803–810. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToidGhvcmF4am5sIjtzOjU6InJlc2lkIjtzOjg6IjcyLzkvODAzIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDYvMTEvMjAyMC4wNi4wMi4yMDExODQ4OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 31. 31.Ewig S, Torres A, El-Ebiary M, et al. Bacterial colonization patterns in mechanically ventilated patients with traumatic and medical head injury. Incidence, risk factors, and association with ventilator-associated pneumonia. Am J Respir Crit Care Med. 1999;159(1):188–198. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1164/ajrccm.159.1.9803097&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9872838&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F11%2F2020.06.02.20118489.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000077987600028&link_type=ISI) 32. 32.Roquilly A, Torres A, Villadangos JA, et al. Pathophysiological role of respiratory dysbiosis in hospital-acquired pneumonia. Lancet Resp Med. 2019;7(8):710–720. 33. 33.Morris AC, Kefala K, Simpson AJ, et al. Evaluation of the effect of diagnostic methodology on the reported incidence of ventilator-associated pneumonia. Thorax. 2009;64(6):516–522. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToidGhvcmF4am5sIjtzOjU6InJlc2lkIjtzOjg6IjY0LzYvNTE2IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDYvMTEvMjAyMC4wNi4wMi4yMDExODQ4OS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 34. 34.Bhat N, O’Brien KL, Karron RA, Driscoll AJ, Murdoch DR, the Pneumonia Methods Working Group. Use and Evaluation of Molecular Diagnostics for Pneumonia Etiology Studies. Clin Infect Dis. 2012;54(suppl 2):S153–S158. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/cid/cir1060&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22403230&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F06%2F11%2F2020.06.02.20118489.atom) 35. 35.Hagel S, Scherag A, Schuierer L, et al. Effect of antiviral therapy on the outcomes of mechanically ventilated patients with herpes simplex virus detected in the respiratory tract: a systematic review and meta-analysis. Crit Care. 2020;24(1):584– 10. 36. 36.Morris AC, Datta D, Shankar-Hari M, et al. Cell-surface signatures of immune dysfunction risk-stratify critically ill patients: INFECT study. Intensive Care Med. 2018;44(5):627–635. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00134-018-5247-0&link_type=DOI) 37. 37.Webber DM, Wallace MA, Burnham CA, Anderson NW. Evaluation of the BioFire FilmArray Pneumonia Panel for Detection of Viral and Bacterial Pathogens in Lower Respiratory Tract Specimens in the Setting of a Tertiary Care Academic Medical Center. J Clin Microbiol. 2020;58:e00343–20 38. 38.Maes M, Higginson E, Pereira-Dias J, et al. Ventilator-associated pneumonia in critically ill patients with COVID-19. Crit Care. 2021;25(1):25–11.