scholarly journals Clinical metagenomics assessments improve diagnosis and outcomes in community-acquired pneumonia

2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Fei Xie ◽  
Zhimei Duan ◽  
Weiqi Zeng ◽  
Shumei Xie ◽  
Mingzhou Xie ◽  
...  

Abstract Background Identifying the causes of community-acquired pneumonia (CAP) is challenging due to the disease’s complex etiology and the limitations of traditional microbiological diagnostic methods. Recent advances in next generation sequencing (NGS)-based metagenomics allow pan-pathogen detection in a single assay, and may have significant advantages over culture-based techniques. Results We conducted a cohort study of 159 CAP patients to assess the diagnostic performance of a clinical metagenomics assay and its impact on clinical management and patient outcomes. When compared to other techniques, clinical metagenomics detected more pathogens in more CAP cases, and identified a substantial number of polymicrobial infections. Moreover, metagenomics results led to changes in or confirmation of clinical management in 35 of 59 cases; these 35 cases also had significantly improved patient outcomes. Conclusions Clinical metagenomics could be a valuable tool for the diagnosis and treatment of CAP. Trial registration Trial registration number with the Chinese Clinical Trial Registry: ChiCTR2100043628.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 144-144
Author(s):  
Vera Grossmann ◽  
Alexander Kohlmann ◽  
Claudia Haferlach ◽  
Hans-Ulrich Klein ◽  
Martin Dugas ◽  
...  

Abstract Abstract 144 PicoTiterPlate (PTP) pyrosequencing allows the detection of low-abundance oncogene aberrations in complex samples even with low tumor content. Here, we compared deep sequencing data of two Next-Generation Sequencing (NGS) assays to detect molecular mutations using a PCR-based strategy and, in addition, to uncover inversions, translocations, and insertions in a targeted sequence enrichment workflow (454 Life Sciences, Roche Diagnostics Corporation, Branford, CT). First, we studied 95 patients (CMML, n=81; AML, n=6; MDS, n=3; MPS, n=3; ET, n=2) using the amplicon approach and investigated seven candidate genes with relevance in oncogenesis of myeloid malignancies: TET2, RUNX1, JAK2, MPL, KRAS, NRAS, and CBL. 43 primer pairs were designed to cover the complete coding regions of TET2, RUNX1 (beta isoform), and hotspot regions of the latter genes. In total, 4128 individual PCR reactions were performed with DNA isolated from bone marrow mononuclear cells, followed by product purification, fluorometric quantitation, and equimolar pooling of the corresponding 43 amplicon products to generate one single sequence library per patient. For sequencing, a 454 8-lane PTP was used applying standard FLX chemistry and representing one patient per lane. The median number of base pairs sequenced per patient was 9.23 Mb. For each amplicon a median of 840 reads was generated (coverage range: 485–1929 reads). As initial proof-of-concept analysis 27 of the 95 patients with known mutations (n=32) as detected by conventional sequencing or melting curve analyses were investigated (range of cells carrying the respective mutation: 1.1% for JAK2 V617F to 98.14% for TET2 C1464X). In all cases, 454 NGS confirmed results from routine diagnostic methods (GS Amplicon Variant Analyzer software version 2.0.01). We then investigated the remaining 69 CMML patients: In median, 2 variances (range 1–8 variances), i.e. differences in comparison to the reference sequence, per patient were detected. These variances included both point mutations in all candidate genes and large deletions (12-19 bp) in CBL, RUNX1, and TET2. Only 20/81 patients of the CMML-cohort (24.69%) were without any detectable mutation. Secondly, in a cohort of six AML bone marrow specimens a custom NimbleGen array (385K format; Madison, WI) was used to perform a targeted DNA sequence enrichment procedure. In total, capture probes spanning 1.91 Mb were designed to represent all coding regions of 92 target genes (1559 exons) with relevance in hematological malignancies (e.g. KIT, NF1, TP53, BCR, ABL1, NPM1, or FLT3). In addition, the complete genomic regions were targeted for RUNX1, CBFB, and MLL. For sequencing, 454 Titanium chemistry was applied, loading three patients per lane on a 2-lane PTP including three molecular identifiers (MIDs) each. Data analysis was performed using the GS Reference Mapper software version 2.0.01. For the enrichment assay, the median enrichment of the targeted genomic loci was 207-fold, as assessed by ligation-mediated LM-PCR. Overall, 1,098,132 reads were generated in the two lanes, yielding a total sequence length of 386,097,740 bases. In median, 96.52% of the sequenced bases mapped against the human genome, and 66.0% were derived from the customized NimbleGen array capture probes, resulting in a median coverage of 18.7-fold . With this method it was possible to detect and confirm point mutations (KIT, FLT3-TKD, and KRAS) and insertions (FLT3-ITD). Moreover, by capturing chimeric DNA fragments and generating reads mapping to both fusion partners this approach detected balanced aberrations, i.e. inv(16)(p13q22) and the translocations t(8;21)(q22;q22) or t(9;11)(p22;q23). In conclusion, both assays to specifically sequence targeted regions with oncogenic relevance on a NGS platform demonstrated promising results and are feasible. The amplicon approach is more suitable for detection of mutations in a routine setting and is ideally suited for large genes such as TET2, ATM, and NF1, which are labor-intensive to sequence conventionally. The array-based capturing assay is characterized by a complex and time-consuming workflow with low-throughput. However, the ability to detect balanced genomic aberrations which are detectable thus far only by cytogenetics and FISH has the potential to become an important diagnostic assay, especially in tumors in which cytogenetics can not be applied successfully. Disclosures: Grossmann: MLL Munich Leukemia Laboratory: Employment. Kohlmann:MLL Munich Leukemia Laboratory: Employment. Haferlach:MLL Munich Leukemia Laboratory: Equity Ownership. Dicker:MLL Munich Leukemia Laboratory: Employment. Kazak:MLL Munich Leukemia Laboratory: Employment. Schindela:MLL Munich Leukemia Laboratory: Employment. Schnittger:MLL Munich Leukemia Laboratory: Equity Ownership. Kern:MLL Munich Leukemia Laboratory: Equity Ownership. Haferlach:MLL Munich Leukemia Laboratory: Equity Ownership.


2017 ◽  
Vol 4 (suppl_1) ◽  
pp. S17-S17
Author(s):  
Krow Ampofo ◽  
Andrew Pavia ◽  
Anne J Blaschke ◽  
Robert Schlaberg

Abstract Background Species-specific polymerase chain reaction (PCR) testing of pleural fluid (PF) from children with parapneumonic effusion (PPE) has increased pathogen identification in pediatric PPE. However, a pathogen is not detected in 25–35% of cases. Hypothesis-free, next-generation sequencing (NGS) provides a more comprehensive alternative and has led to pathogen detection in PCR-negative samples. However, the utility of NGS in the evaluation of PF from children with PPE is unknown. Methods Archived PF (n = 20) from children younger than 18 years with PPE and hospitalized at Primary Children’s Hospital, Utah, in 2015 and previously tested by PCR were evaluated. Ten PCR-negative and 10 PCR-positive PF specimens were tested using RNA-seq at an average depth of 7.7×106 sequencing reads per sample. NGS data were analyzed with Taxonomer. We compared pathogens detected by blood and PF culture, PCR, and NGS. Results Overall, compared with blood/PF culture, PF PCR and PF NGS testing of PF increased bacterial identification from 15% to 50% (P < 0.05) and 65% (P = 0.003), respectively. Pathogen detection in PF by PCR and NGS were comparable (50 vs. 65%, p = NS) (Table). However, compared with PF PCR, NGS significantly increased detection of S. pyogenes (20% vs. 55%; P < 0.05), with 100% concordance when detected by PCR and culture. Detection of Fusobacterium spp. (10 vs. 10%) by PF NGS and PF PCR were comparable. In contrast, there was no detection of S. pneumoniae (15 vs. 0%) by PF NGS compared with PF PCR. Conclusion PF NGS testing significantly improves bacterial identification and comparable to PF PCR testing, which can help inform antimicrobial selection. However there were differences in detection of S. pneumoniae and S. pyogenes. Further studies of NGS testing of PF of children with PPE are needed to assess its potential in the evaluation of PPE in children. Disclosures A. J. Blaschke, BioFire Diagnostics LLC: Collaborator, Have intellectual property in BioFire Diagnostics through the University of Utah and Investigator, Licensing agreement or royalty and Research support; R. Schlaberg, IDbyDNA: Co-founder, Consultant and Shareholder, Stock


Author(s):  
Yi-Yi Qian ◽  
Hong-Yu Wang ◽  
Yang Zhou ◽  
Hao-Cheng Zhang ◽  
Yi-Min Zhu ◽  
...  

Pulmonary infections are among the most common and important infectious diseases due to their high morbidity and mortality, especially in older and immunocompromised individuals. However, due to the limitations in sensitivity and the long turn-around time (TAT) of conventional diagnostic methods, pathogen detection and identification methods for pulmonary infection with greater diagnostic efficiency are urgently needed. In recent years, unbiased metagenomic next generation sequencing (mNGS) has been widely used to detect different types of infectious pathogens, and is especially useful for the detection of rare and newly emergent pathogens, showing better diagnostic performance than traditional methods. There has been limited research exploring the application of mNGS for the diagnosis of pulmonary infections. In this study we evaluated the diagnostic efficiency and clinical impact of mNGS on pulmonary infections. A total of 100 respiratory samples were collected from patients diagnosed with pulmonary infection in Shanghai, China. Conventional methods, including culture and standard polymerase chain reaction (PCR) panel analysis for respiratory tract viruses, and mNGS were used for the pathogen detection in respiratory samples. The difference in the diagnostic yield between conventional methods and mNGS demonstrated that mNGS had higher sensitivity than traditional culture for the detection of pathogenic bacteria and fungi (95% vs 54%; p<0.001). Although mNGS had lower sensitivity than PCR for diagnosing viral infections, it identified 14 viral species that were not detected using conventional methods, including multiple subtypes of human herpesvirus. mNGS detected viruses with a genome coverage >95% and a sequencing depth >100× and provided reliable phylogenetic and epidemiological information. mNGS offered extra benefits, including a shorter TAT. As a complementary approach to conventional methods, mNGS could help improving the identification of respiratory infection agents. We recommend the timely use of mNGS when infection of mixed or rare pathogens is suspected, especially in immunocompromised individuals and or individuals with severe conditions that require urgent treatment.


2019 ◽  
Vol 17 (3.5) ◽  
pp. CLO19-034
Author(s):  
Christie Hancock ◽  
Jacob Bitran

Background: The value of next-generation sequencing (NGS) tests is dependent on how it changes clinical management, if at all. There are not any guidelines on when a physician should obtain testing; what certain genetic variations can be clinically, meaningfully intervened on; and if this active drug is financially practical. A NGS test ideally should identify a targetable mutational profile that leads to a treatment that can be obtained and produces lasting responses. We reviewed the NGS testing practices at our community hospital. Methods: Patients in our oncology clinic that underwent NGS testing of their tumor were reviewed. Specifically, the patient’s age, gender, diagnosis, date of diagnosis, previous lines of treatment, date of NGS testing, and reported molecular targets were recorded. Details of the changes in clinical management based on the NGS and survival time after the NGS test was ordered were also analyzed. Results: Forty-three patient cases were reviewed that had NGS testing obtained between the years 2014–2015 and 2017. Eighteen patients were males and 25 were females. Median age was 60 years. Most common tumor type was breast; 3 were triple-negative and 6 were only hormone-receptor positive. Second most common type was colorectal. NGS changed management in 12 cases. Two patients were able to enter a clinical trial and the other 10 had therapeutic changes based on protein or mRNA overexpression. Median time of change in management to survival in these patients was 7 months. Thirty-seven patients died; overall time from ordering of test to death was 7 months. Overall time of ordering test to survival for all patients was 8 months. Conclusions: NGS gathers a significant amount of information of a patient’s cancer, but this information is difficult to interpret in the clinical setting. Twenty-eight percent of our patients had a change in treatment, but this did not translate into lasting responses compared to other patients. Furthermore, the responses that were seen may have not been related to the therapeutic interventions, but intrinsic tumor biology. It is important to be aware of the clinical utility in ordering these tests for both the physician and patient.


2019 ◽  
Vol 6 (Supplement_2) ◽  
pp. S134-S135
Author(s):  
Andrew D Kerkhoff ◽  
Michelle Matzko ◽  
Charles Chiu ◽  
Steve Miller ◽  
Jennifer M Babik

Abstract Background Many neurologic syndromes are underpinned by infectious etiologies that are difficult to diagnose. Broad-range, universal PCR (uPCR), and metagenomic next-generation sequencing (mNGS) are emerging molecular techniques that may allow for enhanced pathogen detection in challenging cases. To date, their comparative clinical utility for pathogen detection in cerebrospinal fluid (CSF) has not been described. Methods We searched the electronic medical record at University of California, San Francisco for all patients who had mNGS and uPCR results available from the same CSF specimen. Using all available clinical information, patients’ clinical episodes were categorized into one of four categories: (1) confirmed central nervous system (CNS) infection, (2) likely CNS infection, (3) confirmed/likely noninfectious etiology, (4) unknown etiology. We also determined whether mNGS and/or uPCR results changed clinical management. Results We identified 75 patients with 78 paired mNGS and uPCR results on CSF. 14/78 (17.9%) had a confirmed CNS infection underpinning their clinical presentation, 11 (14.1%) had a likely CNS infection, 33 (42.3%) had a likely noninfectious cause, and 20 (25.6%) had etiologies that could not be determined. Of the 14 patients with confirmed CNS infection, n = 4 (28.6%) were diagnosed by mNGS and n = 1 (7.1%) by uPCR (Table 1). Most diagnoses missed by mNGS and uPCR were made by CSF serology or from sites other than CSF. Overall, mNGS detected a pathogen in n = 10/78 (12.8%) cases, compared with n = 4/78 (5.1%) using uPCR (Table 2). Among those with a positive mNGS result, n = 6/10 represented a true or likely true positive result, while the remaining were likely contaminants. Of those with a positive uPCR result, n = 1/4 represented a true positive result, while n = 3/4 were likely contaminants. Clinical management was changed by the mNGS or uPCR result in two cases (Table 2). Conclusion mNGS appears to have superior clinical utility to that of universal PCR for pathogen detection in CSF samples, in large part because of additional ability to detect DNA and RNA viruses. Further studies are required to determine the clinical contexts in which mNGS is likely to have maximal diagnostic yield and to better define the utility of uPCR for CNS infections. Disclosures All authors: No reported disclosures.


2020 ◽  
Vol 21 (S9) ◽  
Author(s):  
Rebecca M. Rodriguez ◽  
Vedbar S. Khadka ◽  
Mark Menor ◽  
Brenda Y. Hernandez ◽  
Youping Deng

AbstractCancer is one of the leading causes of morbidity and mortality in the globe. Microbiological infections account for up to 20% of the total global cancer burden. The human microbiota within each organ system is distinct, and their compositional variation and interactions with the human host have been known to attribute detrimental and beneficial effects on tumor progression. With the advent of next generation sequencing (NGS) technologies, data generated from NGS is being used for pathogen detection in cancer. Numerous bioinformatics computational frameworks have been developed to study viral information from host-sequencing data and can be adapted to bacterial studies. This review highlights existing popular computational frameworks that utilize NGS data as input to decipher microbial composition, which output can predict functional compositional differences with clinically relevant applicability in the development of treatment and prevention strategies.


2019 ◽  
Vol 221 (Supplement_3) ◽  
pp. S331-S340 ◽  
Author(s):  
Augusto Dulanto Chiang ◽  
John P Dekker

Abstract Next-generation sequencing (NGS) technologies have revolutionized multiple areas in the field of infectious diseases, from pathogen discovery to characterization of genes mediating drug resistance. Consequently, there is much anticipation that NGS technologies may be harnessed in the realm of diagnostic methods to complement or replace current culture-based and molecular microbiologic techniques. In this context, much consideration has been given to hypothesis-free, culture-independent tests that can be performed directly on primary clinical samples. The closest realizations of such universal diagnostic methods achieved to date are based on targeted amplicon and unbiased metagenomic shotgun NGS approaches. Depending on the exact details of implementation and analysis, these approaches have the potential to detect viruses, bacteria, fungi, parasites, and archaea, including organisms that were previously undiscovered and those that are uncultivatable. Shotgun metagenomics approaches additionally can provide information on the presence of virulence and resistance genetic elements. While many limitations to the use of NGS in clinical microbiology laboratories are being overcome with decreasing technology costs, expanding curated pathogen sequence databases, and better data analysis tools, there remain many challenges to the routine use and implementation of these methods. This review summarizes recent advances in applications of targeted amplicon and shotgun-based metagenomics approaches to infectious disease diagnostic methods. Technical and conceptual challenges are considered, along with expectations for future applications of these techniques.


2019 ◽  
Author(s):  
Felix Carl Fabian Schmitt ◽  
Anna Lipinski ◽  
Stefan Hofer ◽  
Florian Uhle ◽  
Christian Nusshag ◽  
...  

Abstract Background Patients with sepsis-induced Acute Respiratory Distress Syndrome (ARDS) are hallmarked by high mortality rates. An early and goal directed antibiotic therapy is crucial for patients’ survival. The clinical use of a Next Generation Sequencing (NGS)-based approach for pathogen identification might lead to an improved diagnostic performance. Therefore, the objective of this study was to examine changes in the pulmonary microbiome and resulting influences on patients’ outcome in septic ARDS, but also to compare NGS- and culture-based diagnostic methods for pathogen identification.Results In total, 30 patients in two groups were enrolled in the study: (1) 15 septic ARDS patients and (2) 15 patients undergoing oesophageal resection serving as controls. In the ARDS group, blood samples were collected at ARDS onset as well as 5 days and 10 days afterwards. At the same timepoints, bronchoalveolar lavages (BAL) were performed to collect epithelial lining fluid for culture-, as well as NGS-based analyses and to evaluate longitudinal changes in the pulmonary microbiome. In the control group, only one BAL and one blood sample were collected immediately prior to the surgical procedure. ARDS patients showed a significantly decreased α-diversity (p=0.003**) and an increased dominance (p=0.005**) in their pulmonary microbiome. The α-diversity index revealed a good correlation with the length of stay in the intensive care unit (ICU) (p-value=0.027) and the need for mechanical ventilation (p-value=0.027). In 42.9% of all ARDS patients, culture-based results were not concordant with NGS-based findings. Moreover, culture-based results remained negative in 5 cases where NGS-based diagnostics revealed signs of bacterial colonisation.Conclusion Sepsis-induced ARDS is associated with a significant dysbiosis of patients’ pulmonary microbiome, which is closely correlated with the clinical course of the disease. Furthermore, an NGS-based diagnostic approach was shown to be promising for pathogen identification in septic ARDS.


2021 ◽  
Vol 9 (1) ◽  
Author(s):  
Florian Brandes ◽  
Melanie Borrmann ◽  
Dominik Buschmann ◽  
Agnes S. Meidert ◽  
Marlene Reithmair ◽  
...  

Abstract Background Progranulin is a widely expressed pleiotropic growth factor with a central regulatory effect during the early immune response in sepsis. Progranulin signaling has not been systematically studied and compared between sepsis, community-acquired pneumonia (CAP), COVID-19 pneumonia and a sterile systemic inflammatory response (SIRS). We delineated molecular networks of progranulin signaling by next-generation sequencing (NGS), determined progranulin plasma concentrations and quantified the diagnostic performance of progranulin to differentiate between the above-mentioned disorders using the established biomarkers procalcitonin (PCT), interleukin-6 (IL-6) and C-reactive protein (CRP) for comparison. Methods The diagnostic performance of progranulin was operationalized by calculating AUC and ROC statistics for progranulin and established biomarkers in 241 patients with sepsis, 182 patients with SIRS, 53 patients with CAP, 22 patients with COVID-19 pneumonia and 53 healthy volunteers. miRNAs and mRNAs in blood cells from sepsis patients (n = 7) were characterized by NGS and validated by RT-qPCR in an independent cohort (n = 39) to identify canonical gene networks associated with upregulated progranulin at sepsis onset. Results Plasma concentrations of progranulin (ELISA) in patients with sepsis were 57.5 (42.8–84.9, Q25–Q75) ng/ml and significantly higher than in CAP (38.0, 33.5–41.0 ng/ml, p < 0.001), SIRS (29.0, 25.0–35.0 ng/ml, p < 0.001) and the healthy state (28.7, 25.5–31.7 ng/ml, p < 0.001). Patients with COVID-19 had significantly higher progranulin concentrations than patients with CAP (67.6, 56.6–96.0 vs. 38.0, 33.5–41.0 ng/ml, p < 0.001). The diagnostic performance of progranulin for the differentiation between sepsis vs. SIRS (n = 423) was comparable to that of procalcitonin. AUC was 0.90 (95% CI = 0.87–0.93) for progranulin and 0.92 (CI = 0.88–0.96, p = 0.323) for procalcitonin. Progranulin showed high discriminative power to differentiate bacterial CAP from COVID-19 (sensitivity 0.91, specificity 0.94, AUC 0.91 (CI = 0.8–1.0) and performed significantly better than PCT, IL-6 and CRP. NGS and partial RT-qPCR confirmation revealed a transcriptomic network of immune cells with upregulated progranulin and sortilin transcripts as well as toll-like-receptor 4 and tumor-protein 53, regulated by miR-16 and others. Conclusions Progranulin signaling is elevated during the early antimicrobial response in sepsis and differs significantly between sepsis, CAP, COVID-19 and SIRS. This suggests that progranulin may serve as a novel indicator for the differentiation between these disorders. Trial registration: Clinicaltrials.gov registration number NCT03280576 Registered November 19, 2015.


2020 ◽  
Author(s):  
Manizheh Sayyah-Melli ◽  
Maryam Kazemi-Shishavan ◽  
Nooshin Behravan ◽  
Parvin Mostafa Gharabaghi ◽  
Vahideh Rahmani

Abstract Background: Concurrent bleeding or existing clots usually obscure the vision field and decrease the success rate of hysteroscopy. Therefore, any efforts made in order to have a clear view during the hysteroscopy will improve the diagnostic or treatment outcomes. We examined the effect of preoperative clot evacuation on hysteroscopy and patient outcomes. Methods: In this parallel-group trial, 114 patients with uterine bleeding were randomly assigned to receive either clot evacuation before standard operative hysteroscopy or standard hysteroscopy alone. The investigated endpoints were the clarity of vision, amount of bleeding, the volume of distension media, duration of the procedure, and postoperative complications. Results: All 114 participants completed the study. There were statistically significant differences in the frequency of the clear vision (p<.001), the severity of bleeding (p=.0006), mean procedure time (p<.001), mean serum volume used (p<.001), and the postoperative hematocrit levels (p<.001) between groups, in favor of women with preprocedural intrauterine evacuation. There was no difference related to in-hospital stay (p=0.081) and anesthetic complications among the patients (p=0.182). The procedure was successfully performed on all patients of both groups with zero postoperative complications. Conclusion: Removal of clots and other uterine contents before the insertion of the hysteroscope rendered better and faster access to the uterine wall to observe existing abnormalities. This additional surgical step could take a significant impact on surgical and clinical outcomes. Trial registration: Clinical trial registry name: Iranian Registry of Clinical Trials Url: https://en.irct.ir/trial/33369 The registration number: IRCT20101130005283N13 Date of trial registration: 2018-11-16


Sign in / Sign up

Export Citation Format

Share Document