Skip to main content
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems
  • Log in
  • My alerts
  • My Cart

Main menu

  • Home
  • Articles
    • Latest Articles
    • COVID-19 Special Collection
    • Archive
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Ethics Resources and Policies
  • About the Journal
    • About CMR
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
  • ASM
    • Antimicrobial Agents and Chemotherapy
    • Applied and Environmental Microbiology
    • Clinical Microbiology Reviews
    • Clinical and Vaccine Immunology
    • EcoSal Plus
    • Eukaryotic Cell
    • Infection and Immunity
    • Journal of Bacteriology
    • Journal of Clinical Microbiology
    • Journal of Microbiology & Biology Education
    • Journal of Virology
    • mBio
    • Microbiology and Molecular Biology Reviews
    • Microbiology Resource Announcements
    • Microbiology Spectrum
    • Molecular and Cellular Biology
    • mSphere
    • mSystems

User menu

  • Log in
  • My alerts
  • My Cart

Search

  • Advanced search
Clinical Microbiology Reviews
publisher-logosite-logo

Advanced Search

  • Home
  • Articles
    • Latest Articles
    • COVID-19 Special Collection
    • Archive
  • For Authors
    • Submit a Manuscript
    • Scope
    • Editorial Policy
    • Submission, Review, & Publication Processes
    • Organization and Format
    • Errata, Author Corrections
    • Illustrations and Tables
    • Nomenclature
    • Abbreviations and Conventions
    • Ethics Resources and Policies
  • About the Journal
    • About CMR
    • Editor in Chief
    • Editorial Board
    • For Reviewers
    • For the Media
    • For Librarians
    • For Advertisers
    • Alerts
    • RSS
    • FAQ
  • Subscribe
    • Members
    • Institutions
Review

Emerging Technologies for Molecular Diagnosis of Sepsis

Mridu Sinha, Julietta Jupe, Hannah Mack, Todd P. Coleman, Shelley M. Lawrence, Stephanie I. Fraley
Mridu Sinha
aBioengineering Department, University of California, San Diego, San Diego, California, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Julietta Jupe
bDonald Danforth Plant Science Center, Saint Louis, Missouri, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hannah Mack
aBioengineering Department, University of California, San Diego, San Diego, California, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Todd P. Coleman
aBioengineering Department, University of California, San Diego, San Diego, California, USA
fCenter for Microbiome Innovation, University of California, San Diego, San Diego, California, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Shelley M. Lawrence
cDepartment of Pediatrics, Division of Neonatal-Perinatal Medicine, University of California, San Diego, San Diego, California, USA
dRady Children's Hospital of San Diego, San Diego, California, USA
eClinical Translational Research Institute, University of California, San Diego, San Diego, California, USA
fCenter for Microbiome Innovation, University of California, San Diego, San Diego, California, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Stephanie I. Fraley
aBioengineering Department, University of California, San Diego, San Diego, California, USA
eClinical Translational Research Institute, University of California, San Diego, San Diego, California, USA
fCenter for Microbiome Innovation, University of California, San Diego, San Diego, California, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1128/CMR.00089-17
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

SUMMARY

Rapid and accurate profiling of infection-causing pathogens remains a significant challenge in modern health care. Despite advances in molecular diagnostic techniques, blood culture analysis remains the gold standard for diagnosing sepsis. However, this method is too slow and cumbersome to significantly influence the initial management of patients. The swift initiation of precise and targeted antibiotic therapies depends on the ability of a sepsis diagnostic test to capture clinically relevant organisms along with antimicrobial resistance within 1 to 3 h. The administration of appropriate, narrow-spectrum antibiotics demands that such a test be extremely sensitive with a high negative predictive value. In addition, it should utilize small sample volumes and detect polymicrobial infections and contaminants. All of this must be accomplished with a platform that is easily integrated into the clinical workflow. In this review, we outline the limitations of routine blood culture testing and discuss how emerging sepsis technologies are converging on the characteristics of the ideal sepsis diagnostic test. We include seven molecular technologies that have been validated on clinical blood specimens or mock samples using human blood. In addition, we discuss advances in machine learning technologies that use electronic medical record data to provide contextual evaluation support for clinical decision-making.

INTRODUCTION

Sepsis is a serious and life-threatening clinical condition that generally results from a primary bacterial infection or, less frequently, from a fungal and/or viral infection. Affecting nearly 1 out of every 23 hospitalized patients, it is the sixth most common reason for hospitalization (1–5). At present, it is the most expensive condition treated in U.S. hospitals, with an aggregate cost of US$15.4 billion in 2009 (4, 5), whereas nonspecific diagnoses of sepsis account for another US$23.7 billion each year (6, 7). Alarmingly, the incidence of sepsis is increasing, with a 17% increase in the number of documented cases between 2000 and 2010 (5), while sepsis-related deaths have surged 31% between 1999 and 2014 (8). Approximately 30,000 sepsis-related deaths occur annually, with particularly high rates in critically ill patients admitted to intensive care units (ICUs) (5, 9, 10).

Neonates, or infants within 28 days of life, comprise an additional group at risk for infection due to the relative deficiency of their adaptive immune responses from the lack of antigen exposure in utero as well as the immaturity of innate immune responses, impairments which are directly related to their gestational age at birth. In the United States, sepsis is the fifth leading cause of neonatal mortality, surpassed only by loss of life due to preterm birth and intrapartum complications (11–13). Furthermore, infection has been linked to preterm birth (14–16). Devastatingly, 25% of all neonates in the United States admitted to a neonatal ICU (NICU) will be diagnosed with sepsis, and 18 to 35% (21,000 neonates/year) will die from their infection (11, 17, 18). Low-birth-weight premature infants have a 10-fold increased risk of serious infection or sepsis compared to their full-term counterparts, with a 30% mortality rate (19–21).

Septic patients usually present with malaise, fever, chills, and leukocytosis, which often prompt care providers to evaluate patients for the presence of bacteria in the bloodstream (bacteremia) by using blood culture analysis. Considered a medical emergency, sepsis can rapidly progress to organ dysfunction and death despite immediate and aggressive medical therapies (10). In the absence of robust diagnostic tests, the reflexive utilization of broad-spectrum and highly potent antibiotic treatment in patients suspected of having sepsis has contributed to the emergence of drug-resistant organisms and atypical pathogens (22, 23). Survivors of sepsis may experience substantial long-term complications leading to a prolonged length of stay and/or discharge to a long-term-care setting (6). Neonatal sepsis survivors are at an increased risk for poor neurodevelopmental outcomes, including cerebral palsy, deafness, blindness, and cognitive delays (11, 24).

Because of the high mortality rate associated with sepsis, the dangers of undertreating some infections, or concerns about the use of inappropriate antibiotics, physicians tend to order blood cultures liberally (10). This results in bacteria being isolated in only 4 to 12% of processed blood culture tests. In more-restrictive settings where blood cultures are ordered less liberally, positivity rates can be much higher. Regardless, the detection of positivity occurs several hours to days after the patient has been treated (25–29).

Pathogen detection by blood culture is, unfortunately, worse in the neonatal patient population than in older children and adults. A major confounding factor arises from the fact that clinical signs related to sepsis are similar to those of other noninfectious life-threatening conditions, such as perinatal asphyxia, respiratory distress syndrome, and symptoms associated with severe prematurity. Although more than 60% of sepsis evaluations occur in the first 3 days of life, <1% of blood culture tests detect an organism. Even in symptomatic neonates, blood culture methodologies can detect the offending microorganism in only 10 to 15% of neonates after contaminants are excluded (30, 31). This burden is worse in underserved communities. For example, in the United States, black preterm neonates have the highest incidence of and case fatality rate from neonatal sepsis (32). Around the world, neonates born in low- and middle-income countries suffer the highest rates of sepsis (33). Critically, in low- and middle-income countries, resistant bacterial strains are implicated in the majority of the cases, highlighting the need for rapid susceptibility testing.

Underrecognition of illness in addition to the emergence of resistant pathogens, delays in diagnosis, and the inability to access or afford specialized medical care contribute to the high mortality and morbidity rates associated with sepsis (34). The correct initial choice of antibiotic therapy has been shown to save more lives than any other medical intervention (35–38), and studies suggest that there is a 1- to 3-h diagnostic window, from symptom-based sepsis recognition to the initiation of antimicrobial treatment, before the mortality rate increases (39). The Surviving Sepsis campaign advocates for the implementation of antibiotic therapy within 1 h of clinical recognition of sepsis and collection of blood for culture prior to the administration of antibiotics (35). However, a 5-fold reduction in survival has been reported as a consequence of inappropriate antimicrobial therapy in the first 6 h after the recognition of sepsis (40). A recent editorial questioned blind antibiotic use and made a compelling case for the initiation of targeted antimicrobial therapy, preferably after the detection of the pathogen (41). Thus, rapid diagnostic tests that are capable of profiling antimicrobial resistance or ruling out bacterial infection as a cause of sepsis must be integrated into the initial 1 to 3 h of the clinical timeline to influence the appropriate use of antibiotics and patient outcomes.

Unfortunately, findings from standard diagnostic tests are not available within this critical time frame to allow focused, effective, and potentially life-saving medical interventions. Other faster adjunct standard hematological analyses used in routine clinical practice have low sensitivity and specificity, particularly in neonatal patients (42). Recently, biomarkers such as C-reactive protein (CRP), procalcitonin (PCT), and the neutrophil marker CD64 have made their way into sepsis evaluations, with limited success. Most diagnostic approaches that are currently employed rely on individual biomarkers, with binary yes or no answers. An integrative diagnostic strategy that incorporates a broader range of biomarkers could in theory characterize the host response to rule in/out infection, identify and quantify the pathogen(s), and predict resistance. Such a test is greatly needed to distinguish patients who are truly septic and assist with the appropriate use of antibiotics.

THE IDEAL SEPSIS DIAGNOSTIC TEST

Considering the current clinical challenges and the need to impact clinical management by informing targeted treatment, the ideal technology should include the following characteristics (43, 44):

  1. rapid detection (the pathogen needs to be identified in less than 3 h) (35, 39);

  2. broad-based detection, including bacteria, viruses, and fungi;

  3. minimal invasiveness, utilizing clinical samples with low specimen volumes (<1 ml blood for pediatric patients, including neonates, and 5 to 10 ml blood for adults) (45–47);

  4. high sensitivity and specificity for the immediate initiation of targeted antibiotic use in the presence of signs and symptoms of systemic inflammation (the diagnostic tests should not compromise on sensitivity with low pathogen levels in the specimen);

  5. polymicrobial detection of pathogens in the presence of contaminants across a wide range of pathogen loads (∼1 to 100,000 CFU/ml blood);

  6. detection of drug resistance;

  7. integration into the clinical workflow (the process should be easy to use and require minimal technical expertise to process samples and interpret test results; for the greatest impact, the technology must be usable in noncentralized low-resource settings);

  8. the ability to detect unknown and emerging pathogens (detection capabilities must be able to easily expand without compromising the robustness of detection and the required specimen volume); and

  9. the ability to distinguish the inflammatory response as being either host or pathogen driven (48, 49).

LIMITATIONS OF STANDARD DIAGNOSTIC BLOOD CULTURE METHODOLOGIES (“GOLD STANDARD”)

Today, the use of standard culture techniques for the detection and isolation of pathogenic organisms from a sterile body fluid specimen is still considered the “gold standard” for the diagnosis of infection and sepsis (50). However, routine blood cultures can take 6 h to 5 days to grow an organism to detectable levels, with additional time being required to identify (24 h) and test for (48 h) antibiotic susceptibility (28, 51, 52). This test is also plagued by many complicating factors. First, the quantity of microbes present in circulation during bloodstream infection (BSI) is usually low, ranging from only 1 to 1 × 104 CFU/ml (24, 53–55). In older children and adults, routine blood culture tests are performed in timed sequences of up to four separate replicates comprising approximately 20 to 30 ml of blood each. This repeat blood sampling improves pathogen detection to capture the causative organism in 73 to 95% of cases (35, 55–58). Small sample volumes can therefore lead to false-negative results with conventional practices (59–61). Unfortunately, in neonates, especially very-low-birth-weight (VLBW) (birth weight of <1,500 g) premature infants, blood collection is restricted to a single sample with a minimal volume (1 ml), which can further hinder pathogen capture, particularly when the level of bacteremia is low (45–47). Neonatal sepsis concentrations often fall within the range of 1 to 1,000 CFU per ml, with some studies finding that concentrations in 68% of culture-positive cases fall below 10 CFU per ml (62, 63).

False-negative results can also occur due to the challenge of recovering infectious etiologies by routine blood culture techniques after the initiation of antibiotic therapy, which affects 28 to 63% of adults with suspected sepsis (35, 55, 61, 64, 65). Exposure to antimicrobials prior to blood culture testing is magnified in neonatal patients, as an estimated 30 to 35% of laboring women receive empirical intrapartum antibiotics for the prevention of neonatal group B Streptococcus (GBS) disease (21). Subsequently, compliance with Centers for Disease Control and Prevention (CDC) GBS guidelines exposes an estimated 65% of VLWB infants to antibiotics prior to birth (66–68). Prolonged delays in pathogen identification and antibiotic susceptibility testing also cause neonates to be unnecessarily exposed to broad-spectrum antibiotics, leading to bacterial antibiotic resistance in noninfected neonates while preventing targeted antimicrobial therapy in septic neonates. Additionally, prolonged broad-spectrum antibiotic exposure in neonates can lead to invasive fungal (Candida) infection, necrotizing enterocolitis, and death (17, 18, 69).

Failure to adhere to standard antiseptic procedures during sample collection can also lead to contaminated, or false-positive, blood culture results. In 2005, the College of American Pathologists reported an overall mean blood culture contamination rate of 2.89% in 356 institutions, with rates of 2.08% noted for neonatal patients and 2.92% noted for nonneonatal patients (70). Contamination rates for individual institutions in this study ranged from 2.15% to 3.67% and contributed to an additional estimated cost of US$5,506 per patient (70). Thus, contaminated samples can have enormous financial and clinical ramifications in adult populations in the United States, including 1,372 to 2,200 extra hospital days and an extra US$1.8 million to US$1.9 million in medical costs each year (71, 72). In pediatric patients, these tainted samples are associated with readmission rates of 14 to 26% (61, 73, 74) and increased lengths of stay from 1 to 5.4 days (61, 72, 75). In low- and middle-income countries, where there is a dearth of trained medical staff and quality health care services, blood culture contamination is not uncommon and can have grave consequences. Notably, almost half of patients with false-positive blood cultures are treated with antimicrobials compared to those with true-positive test results (61, 76–78). Additionally, 40 to 50% of adult patients with bacteremia (and 70% with fungemia) received incorrect antimicrobial therapy during their empirical treatment period before microbiology culture results were available (1, 5, 79). This misuse of antimicrobial agents and delays in pathogen identification cause prolonged exposure to broad-spectrum antibiotics, which can also result in an increased number of Clostridium difficile infections, antibiotic allergic reactions and drug toxicity, antimicrobial-resistant bacterial strains, prolonged length of stay, and increased medical costs (5, 61, 80–82). Additional approaches to mitigate contamination have been described with some success, such as algorithms, including clinical judgments, numbers of positive blood culture sets among all sets obtained, and ancillary laboratory tests such as CRP and PCT measurements (83).

In summary, routinely used blood culture methods are not an ideal gold standard, as the results often come too late, are incomplete or not sensitive enough, and can be misleading and relatively labor-intensive. There is a crucial unmet need to shorten as well as improve current laboratory procedures for the detection and identification of microorganisms. In the last decade, various engineering innovations have generated promising pathogen detection approaches that incorporate sample preparation, molecular detection, automation, miniaturization, multiplexing, and high-throughput analysis toward the development of an effective diagnosis technology. The following sections give an overview of current and emerging detection systems designed for the rapid, sensitive, and cost-effective diagnosis of bloodstream infections.

TOWARDS DETECTION DIRECTLY FROM WHOLE BLOOD: CURRENT AND EMERGING TECHNOLOGIES FOR RAPID DIAGNOSIS OF MICROBIAL INFECTIONS WITHOUT CULTURE

In the United States today, nearly all U.S. Food and Drug Administration (FDA)-approved sepsis molecular diagnostic tests are postculture technologies, meaning that a blood sample must be cultured to allow the number of microbes to increase before the diagnostic test can be conducted. This initial growth-based amplification ensures sensitive detection but extends the diagnostic timeline such that test results do not effectively impact patient management. It also restricts the breadth of organisms detected by relying on a single culture medium formulation, which cannot support the growth of all organisms or may mask susceptibilities (84–87). While molecular diagnostic tests are completed within 20 min to 2 h, the initial step of blood culture takes several hours to days and may not be successful. Likewise, determining the antibiotic susceptibility of the pathogen also depends first on additional culture methods. Current technologies do not benefit antibiotic stewardship programs aimed at deescalating empirical antibiotic therapy and encouraging timely targeted treatment. Recent reviews by Opota et al. (55, 88), Kothari et al. (89), Afshari et al. (90), and Ecker et al. (91) describe the state of the art for such diagnostic tests in more detail. In this review, we focus on emerging technologies that are not dependent upon initial microbial growth. All technologies described in the following paragraphs are summarized in Table 1.

View this table:
  • View inline
  • View popup
TABLE 1

Emerging molecular diagnostics for pathogen detection from whole blooda

Modern Nucleic Acid Amplification TechnologiesFor several years, nucleic acid amplification technologies (NAATs) have promised to circumvent the need for bacterial growth. These technologies function by rapidly creating copies of DNA or RNA originating from pathogen or host cells through biochemical reactions, amplifying the nucleic acid sequences to a detectable level. The sequences are then used to identify the infecting agent or the status of the immune response. However, the promise of NAATs for revolutionizing sepsis diagnosis has yet to be realized. This can be attributed to challenges in reliably capturing and amplifying pathogen nucleic acids from complex samples like blood, where the infecting agents are present at low levels or as polymicrobial mixtures within a high background of human DNA. In this sample context, traditional NAATs cannot simultaneously satisfy the need for sensitive, specific, and broad-based detection. The emerging technologies discussed here represent novel integrations of NAATs with other cutting-edge techniques that together are capable of overcoming many current diagnostic limitations. We also discuss the exciting promise that further synergistic integration holds for producing the ideal sepsis diagnostic test.

Iridica Plex ID.The Iridica Plex ID platform (Abbott Molecular, Des Plaines, IL) boasts the most broad-based detection of any direct-from-blood emerging technology, identifying an impressive 780 bacteria and Candida species with a turnaround time of 6 h (55, 92). However, only four antimicrobial resistance markers (mecA, vanA, vanB, and blaKPC) are detected. Iridica accomplishes this by integrating multiplexed PCR amplification of pathogen DNA with electrospray ionization mass spectrometry (ESI-MS) for sequence identification. The process starts with automated DNA extraction from a 5-ml whole-blood sample. The extracted DNA is distributed across several PCR mixtures containing different primers targeting conserved regions of pathogen genomes, including the 16S and 23S rRNA genes for bacteria and Candida, respectively. These primers and reaction components have been optimized to limit interference due to human DNA, which can otherwise lead to nonspecific amplification or low amplification efficiency. Amplified copies from each reaction are selectively enriched by removing over 98% of human DNA. They are then assessed by ESI-MS, which generates nucleotide base composition data. Finally, the data from each amplicon are compared to a library of all expected base compositions and used to triangulate the pathogen species (Fig. 1) (54, 93).

FIG 1
  • Open in new tab
  • Download powerpoint
FIG 1

Workflow for the analysis of a single whole-blood specimen for pathogen identification. Even though U-dHRM shows promise as the fastest technology, it could benefit from parallelizing for multiple loads in the future.

While this approach achieves a wide breadth of detection, clinical studies show that the sensitivity, specificity, and negative predictive value (NPV) of Iridica vary widely from 45% to 83%, 69% to 94%, and 80% to 97%, respectively, against conventional culture methods (Table 2 and Fig. 2). After the exclusion of possible contaminant bacteria and estimation of true-positive rates based on PCR test replicates or clinical chart and culture results for patient-matched specimens, sensitivity and specificity values can be improved from 77% to 91% and from 87% to 99%, respectively (54, 92). Improvement with multiple test replicates suggests that sample heterogeneity and sampling error remain problematic. In the case of a low level of a pathogen, sampling error first occurs at the point of the blood draw, is combined with any inefficiency in nucleic acid extraction, and then occurs again when the sample is split across multiple distinct amplification reactions targeting different genes on the Iridica platform. Improving the sensitivity and reliability of detection will require circumventing these sources of error. For the same reasons plus amplification competition, polymicrobial samples may present another challenge for this approach. Some evidence suggests that the Iridica platform can detect mixed pathogen populations, but its utility for clinical samples is currently inconclusive. We found only one study that investigated polymicrobial specimens. Here, the Iridica platform identified only one causative organism in four out of nine cases of blood culture-positive polymicrobial infection (94).

View this table:
  • View inline
  • View popup
TABLE 2

Characteristics of studies reviewed for data on the Iridica platform

FIG 2
  • Open in new tab
  • Download powerpoint
FIG 2

Sensitivity plotted against specificity of test results compared against the gold standard of blood culture for Iridica, SeptiFast, and SepsiTest. The marker/symbol area is proportional to the number of paired blood tests in the study. Darker shades of color signify larger blood volumes used for the test. (A) For Iridica, we included data from 6 publications found by a PubMed literature search. (B) For SeptiFast, we included data from 2 meta-analyses (summary statistics from analyses are shown in black, along with the confidence intervals) in addition to data from 8 new relevant studies. (C) For SepsiTest, we included data from 5 publications found by a PubMed literature search.

Iridica has been evaluated in a limited number of clinical studies across patients with suspected sepsis, systematic inflammatory respiratory syndrome (SIRS), and febrile neutropenia (54, 92–95). Interestingly, significant differences in sensitivity have been reported across ICU and emergency room (ER) patients (P = 0.005), with higher sensitivity being seen for ICU patients (92). This may derive from higher pathogen loads in this patient population, which would have the effect of reducing sampling error. Limits of detection of the Iridica platform range from 0.25 to 128 CFU/ml for bacteria, depending on the target species, and 4 CFU/ml for Candida species (54, 93).

This broad-based semiquantitative technology shows promise for use on whole-blood samples to detect a wide variety of pathogens, but its potential impact on antibiotic stewardship is low given the limited number of resistance markers detected. However, this technology benefits from the ability to expand the test to more targets in the future. The use of 5 ml blood is promising for adult patients but limits feasibility for use on pediatric patients (96). The Iridica platform is an end-to-end diagnostic solution with a structured and easy-to-use workflow. Individual steps are automated, thus reducing labor and increasing efficiency. The time to detection ranges from between 6 and 8 h with only 30 min of hands-on time for a batch of 6 samples (93, 94). However, this technology fails to meet the ideal turnaround time of 1 to 3 h. This technology is not yet approved by the U.S. FDA but is Conformité Européenne (CE) marked, meaning that it complies with the European In-Vitro Diagnostic Devices Directive and is commercially available in Europe (55, 91). However, it may fall short in noncentralized clinical settings due to the dependence on multiple bulky devices and high up-front costs of about US$357,043 (90, 97). The cost per test ranges from US$262.92 to US$419.14 (assuming that £1 equals US$1.33) (98).

SeptiFast.SeptiFast (Roche Diagnostics, Risch-Rotkreuz, Switzerland) is a commercially available (in the European Union), broad-based microbe identification test for whole blood. It can identify over 16 bacteria, 5 Candida species, and Aspergillus fumigatus fungi using a 1.5-ml whole-blood sample within 6 h. In addition, it can detect the mecA antibiotic resistance gene after a sample tests positive for Staphylococcus aureus. This technology is CE marked but not yet FDA approved.

The SeptiFast test integrates multiplexed real-time PCR with probe hybridization and DNA melting analysis. The test begins with nucleic acid extraction from whole blood under a contamination-controlled workflow. This is followed by real-time PCR amplification using a combination of universal and specific primers in three parallel reactions for Gram-positive bacteria, Gram-negative bacteria, and fungi (99). The primers target the internal transcribed spacer (ITS) regions between the 16S and 23S genes for bacteria and between the 18S and 5.8S genes for fungi. PCR products are detected by using species-specific probes that fluoresce in one of the four detection channels. Species identified in the same detection channel are subsequently differentiated by using melting temperature analysis (Fig. 1) (99, 100).

SeptiFast has a reported sensitivity of between 3 and 100 CFU per ml, depending on the microorganism (99). A meta-analysis of 41 studies reported a summary sensitivity and specificity of 68% (95% confidence interval [CI], 63% to 73%) and 86% (95% CI, 84% to 89%), respectively, for a total of 10,493 SeptiFast tests compared to blood culture (101). Another meta-analysis that included only data from journal publications reported slightly better overall sensitivity and specificity, 75% (95% CI, 65% to 83%) and 92% (95% CI, 90% to 95%), respectively, based on 8,438 tests (102). Recent studies show similar heterogeneous results (Table 3 and Fig. 2) (100, 103–140). These numbers improved when studies incorporated clinical markers along with blood culture results (124, 125, 137, 141). However, as many as 35% of the SeptiFast-positive episodes were not supported by any microbiological or clinical data (109, 120). On the other hand, low sensitivity prevented SeptiFast from identifying culture-positive organisms in 20 to 30% of cases (142). SeptiFast has been reported to resolve polymicrobial infections with higher detection rates (χ2 = 4.50; P = 0.0339) than blood culture (121, 132, 133, 143, 144). However, the detection of mixed pathogens may be hindered by competing amplification due to the use of multiple specific primers and needs further investigation (137).

View this table:
  • View inline
  • View popup
TABLE 3

Characteristics of studies reviewed for data on SeptiFasta

In summary, SeptiFast may be considered broad-based, with coverage of the 25 most relevant pathogens for sepsis, and incorporates the ability to detect mixed pathogen populations. However, it is missing pathogens that are highly relevant for neonatal sepsis. This technology considerably lowers the blood volume needed for testing compared to that needed for conventional technologies, which could be beneficial for pediatric patients (100). However, 1.5 ml of blood is excessive for neonates, for whom samples are limited to 1 ml. SeptiFast, when used with MagNA Pure (Roche) automated DNA extraction, shortens the complete workflow time to 3.57 h for eight parallel loads (115). This diagnostic test may be of added value for the management of patients with suspected sepsis who are SeptiFast positive but blood culture negative (106, 121, 129, 145, 146). However, low sensitivity deems negative results nonactionable. It may also fall short in noncentralized clinical settings due to the dependence on multiple bulky devices and up-front costs of about US$35,167 (90, 97). The cost per test is estimated to range from US$204.73 to US$273.83 (assuming that £1 equals US$1.33) (98). Other limitations include incomplete antibiotic resistance information and the inability to expand the test due to a limited number of detection channels.

SepsiTest.SepsiTest (Molzyme, Bremen, Germany) is a commercially available (in the European Union) broad-based microbial identification test for whole blood. It can identify over 345 bacteria and 13 fungi in 8 to 10 h from a 1-ml whole-blood sample. This technology is CE marked and commercially available in Europe but not yet FDA approved.

SepsiTest integrates universal PCR with Sanger sequencing after a unique sample preparation step whereby selective lysis and human DNA degradation are used to improve sensitivity (147). After DNA is isolated, PCR is performed with a universal primer targeting the 16S and 18S rRNA genes for bacteria and fungi, respectively. Bacteremia or fungemia is reported in <4 h. Further purification followed by Sanger sequencing accomplishes species detection, which takes an additional 4 to 6 h (Fig. 1).

SepsiTest can detect as few as 10 to 80 CFU/ml, with some organism bias (148, 149). It has a reported sensitivity ranging from 11% to 87% and a high specificity ranging from 85% to 96% compared to blood culture in adult and pediatric patients with SIRS, sepsis, febrile neutropenia, and infectious endocarditis (130, 150–154) (Table 4 and Fig. 2). Multiple studies report promising NPVs close to 97% against blood culture with the detection of multiple fastidious organisms (152, 154). Similar sensitivities ranging from 37.5% to 78.6% and specificities ranging from 86.8% to 94.4% were observed in studies adjusting for clinical context by excluding contaminants (130, 153). Additionally, as many as 45% of PCR-positive test results were reported to be due to contaminants (153). Pathogens detected in mixed populations were often identified as contaminants (151, 152). In one reported study, only one organism was identified in three of four blood culture-positive polymicrobial specimens (152).

View this table:
  • View inline
  • View popup
TABLE 4

Characteristics of studies reviewed for data on SepsiTesta

SepsiTest is a broad-based test that requires a small amount of blood appropriate for both adult and pediatric patients. It can, in principle, detect polymicrobial infections; however, its ability to inform clinical decision-making needs further study. SepsiTest provides the option to automate DNA extraction (SelectNA plus; Molzyme) and process up to 12 samples in one run, making it easy to integrate into the clinical workflow. However, it does not provide any information on antibiotic sensitivity. In addition, it still requires multiple steps that are not integrated into one platform, increasing the risk of contamination and the turnaround time. This limits its utility for informing clinical decisions regarding targeted antimicrobial therapy. It may also fall short in noncentralized clinical settings due to the dependence on multiple bulky devices and high up-front costs of about US$57,553 (90, 97). The cost per test ranges from US$144.28 to US$199.21 (assuming that £1 equals US$1.33) (98). The use of Sanger sequencing is the time-limiting step for SepsiTest. In the future, massively parallelized next-generation sequencing technologies may enhance this approach and provide antibiotic resistance information. In the next paragraph, we provide a short summary of such an emerging sequencing technology.

Nanopore sequencing (MinION).The MinION (Oxford Nanopore Technologies, Oxford, United Kingdom) is a portable, real-time, USB-powered DNA/RNA sequencer with a 10- to 50-min library preparation step. The main advantages of the MinION platform over other next-generation sequencing technologies are its (i) rapid turnaround, (ii) low capital cost, and (iii) small size. This technology was released to researchers for alpha testing as part of an early-access program in 2014 (155). It is a generic sequencing system that has shown the potential for the rapid identification of pathogens (<4 h) (156) directly from blood when combined with a PCR amplification step using the 16S Rapid Amplicon Sequencing kit (155). Because it performs sequencing at the single-molecule level, it offers new possibilities to study microbial diversity in clinical samples and also allows multiplexing of samples. This technology has been validated for viral pathogen identification from 140 μl whole blood in <40 min with 100% sensitivity and specificity (157, 158). For bacteria, it has been validated only with clinical urine and fecal samples (156, 158). Polymicrobial pathogen identification has been demonstrated by using genomic DNA mixtures of 20 bacterial strains in equal amounts (100,000 copies per organism per μl) (159, 160). By using specific primers that amplify a wide range of bacterial 16S rRNA genes, 90% of the full-length 16S rRNA gene could be reconstructed with the MinION nanopore technology. However, pathogen assignment could be completed for only 8 of the pathogens from the DNA mixture due to low sequencing coverage. This was attributed to nonoptimized 16S PCR amplification, despite the use of universal primers (159). This points to the need for the optimization and validation of this technology as a complete system with whole blood. Other improvements are needed to transition MinION into the clinic. These improvements include automation; standardized external and internal spike-in controls that run in parallel to prevent carryover contamination (161); as well as optimization of the bioinformatic pipeline used to identify organisms, resistance genes, and/or mutations (156, 162, 163). A major benefit of this technology is that it does not require up-front capital costs. According to the Oxford Nanopore Technology website, it is currently being offered to members of a developer-style access program for US$1,000, which includes the MinION device, three flow cells, two reagent kits, and software.

U-dHRM and machine learning on pathogen DNA fingerprints.The universal digital high-resolution melt (U-dHRM) platform is a broad-based microbial identification technology used with whole-blood samples. It can currently detect 37 bacterial pathogens with single-organism and single-genome sensitivity as well as resolve polymicrobial infections in <4 h using <1 ml whole blood (164, 165). This technology is in the validation phase (University of California, San Diego) and is not yet commercially available.

U-dHRM integrates universal digital PCR (dPCR) with high-resolution melt (HRM) analysis on a microfluidic chip to enable probe-free differentiation and quantification of bacteria within a sample (165). The test procedure begins with DNA extraction followed by sample “digitization,” which separates all pathogen genomes into their own PCR mixtures by spreading the sample across a microfluidic chip containing 20,000 picoliter-sized reaction mixtures. In each reaction, universal amplification targeting the 16S rRNA gene takes place. Subsequently, precise heating and simultaneous imaging are performed on all reaction mixtures to generate HRM curve fingerprints for each pathogen's 16S gene sequence (Fig. 1). HRM generates sequence-specific melt curves by unwinding DNA amplicons in the presence of a fluorescent double-stranded intercalating dye (166–169). Each distinct DNA sequence melts uniquely, generating a loss of the fluorescence signature as a function of temperature that is then used for species identification (Fig. 3). A supervised machine learning algorithm automatically identifies the microbial species by its melt curve. U-dHRM has reported a classification accuracy of 99.9% for the 37 pathogens tested, with load quantification for individual pathogens (165). This technology was validated by using mock blood samples, demonstrating its ability to identify pathogens in the presence of excessive human DNA (165).

FIG 3
  • Open in new tab
  • Download powerpoint
FIG 3

Digitization and melting of genomes after amplification with PCR technology. A melt curve corresponding to the individual genome is generated for identification and absolute load quantification of the pathogen and contaminants.

U-dHRM is a rapid, broad-based test to detect multiple organisms in a blood sample of less than 1 ml, which is suited for pediatric patients and neonates. While it is currently limited to 37 bacteria relevant to neonatal sepsis, it has the potential to expand to include additional bacteria, fungi, and viruses in the future. Since this technology is probe free and digitized, it has the potential to detect all sepsis-causing organisms contained in a single sample, including polymicrobial infections. Early studies showed promising single-genome sensitivity and 99.9% specificity, but further evaluation with clinical blood samples is needed. Validation at other sites will also be important. In addition, automation is required to accomplish a sample-to-answer time of <3 h. This system is easy to use and can incorporate the detection of antibiotic resistance determinants. Its machine learning framework provides the potential identification of new and unknown pathogens and allows for an expanding library. The speed and simplicity of U-dHRM along with its integrated technology platform suggest a promising first-pass screening method for neonatal sepsis. This technology also shows the potential to deliver at- or near-point-of-care diagnosis. The possibility of moving U-dHRM toward a portable, inexpensive system can be of immense value to noncentralized systems in low-resource settings (170, 171), where the cost per test must typically fall between US$1 and US$30.

Summary of modern nucleic acid amplification technologies.In summary, the results of clinical studies using PCR-based technologies are heterogeneous. For the most part, these results are reported in comparison with the gold standard of blood culture, which is far from ideal and may contribute significantly to this heterogeneity. Blood may be drawn with varied timing, at different bodily locations, and in various amounts for blood culture. This contributes to the challenge of validating emerging technologies against blood culture. Hence, it is important to interpret diagnostic results in conjunction with clinical context.

(i) Interpreting false-positive results against blood culture.One of the major advantages of a PCR-based technology is its ability to detect nonviable, fastidious, and unculturable organisms that would otherwise be missed by blood culture. A PCR-positive, blood culture-negative specimen may reflect a real pathogen yet leads to biased lower sensitivity and specificity values of the PCR test. Hence, it is crucial that we carefully design comparison studies to include other molecular and adjunct tests instead of relying solely on an imperfect gold standard. It should be noted that false-positive results could also be due to cell-free pathogen DNA circulating in the blood originating from an old or controlled infection or contamination (172). Typically, PCR-based methods are unable to differentiate between viable and nonviable DNA (173). On the contrary, the use of 16S gene primers to amplify long 1-kb amplicons by U-dHRM may allow the differentiation of intact DNA relevant to active infection from the degraded DNA of dead pathogens or from the environment (165, 174). This can be a significant advantage over other molecular diagnostic tests, including sequencing technologies, which detect a high background level of organisms; clinical trials are pending. It has been reported that both Iridica and SepsiTest have higher rates of contamination than blood culture (95, 153), which likely arises from the use of broad-based universal primers that are targeted to amplify short fragments of DNA. In addition, as shown in Fig. 2, SepsiTest and Iridica involve more sample transfer steps than SeptiFast and U-dHRM. This further increases the risk of contamination (153).

Both SeptiFast and Iridica use semiquantitative methods to detect contaminants and limit false-positive results. SeptiFast uses a cutoff value that represents the number of PCR cycles at which DNA is adequately amplified to identify contaminants (175, 176). Iridica also uses similar thresholds based on the number of genomes per well to limit contaminants and reduce false-positive results. However, these techniques may need further optimization, as they can conversely lead to false-negative results (92, 176). Absolute load quantification in conjunction with clinical characteristics may improve diagnostic accuracy as well (175). An emerging theme is the need for integrating quantitative results with clinical context, potentially provided by a machine learning framework. For example, a diagnostic algorithm that uses a patient's CD64 index to determine whether SepsiTest should be performed has been proposed. This approach showed an improved detection of pathogens in patients with suspected BSI (177). A similar approach in conjunction with neutrophil/lymphocyte count ratios and levels of presepsin and procalcitonin has been suggested (150, 178).

U-dHRM manages contamination through the use of small reaction mixture volumes, which keeps contaminants from overwhelming low-level pathogen DNA in the amplification step. This also enables absolute quantification, since each organism's genome is amplified individually, without affecting detection sensitivity (164). It also integrates the amplification (dPCR) and detection (HRM) steps into a single closed system, which eliminates contamination due to sample transfer and reduces hands-on time (164). Importantly, the ease of use, speed, and quantitative power of this technology could enable repeated testing to track the appearance and removal of bacterial DNA in the blood during antibiotic treatment. In combination with host inflammatory markers, such repeated testing could lend deeper insights into the progression of sepsis. Having the ability to conduct repeated testing over time could reveal novel disease dynamics that may contribute to a further understanding of pathogen detection inconsistencies that often arise in technology comparison studies. U-dHRM also holds promise to address the need for point-of-care diagnostic tests, whereas other commercially available PCR tests typically need bulky and expensive equipment that is not feasible for use in noncentralized systems.

(ii) Interpreting false-negative results against blood culture.While false-positive results may lead to the inaccurate overuse of antibiotics and contribute to the generation of resistant organisms, false-negative results and the inaccurate withholding of antibiotic treatment are more immediately threatening to patient welfare (179). Accurately withholding empirical antibiotic use will require an improved sensitivity of PCR technologies (>98% negative predictive value) (43). PCR tests can be limited in their ability to detect pathogens for a variety of reasons, including the need for effective lysis across a broad range of microbes, the interference of human DNA or other inhibitory substances carried over from blood into the assay mixtures, the effect of off-target interactions, and amplification bias (180–182). It is interesting to note that even though all the above-described technologies rely on an initial PCR amplification step for microbe detection followed by a secondary step for species identification, they differ in their diagnostic sensitivities. The two major contributors to these differences are (i) the approach used for reducing interference from human DNA and (ii) the amplification strategy using either a single universal primer (SeptiTest and U-dHRM) or multiple broad-range or species-specific primers (Iridica and SeptiFast), which may suffer from amplification competition (Fig. 1). All commercially available PCR tests have optimized their workflow to improve pathogen DNA amplification, yet none of them show promise to replace blood culture due to their limited sensitivity for clinical specimens (Fig. 2). The Iridica platform recently increased the sample volume 5-fold, from 1 ml to 5 ml, under the assumption that low sensitivity arises from the inefficient capture of the pathogen in low-volume blood samples (54).

The enhanced sensitivity of U-dHRM is attributed to the diluting effect of the digital reaction format on inhibitory substances and the optimized dPCR reaction conditions ensuring the amplification of single copies of bacterial DNA. U-dHRM has been shown to significantly reduce false-negative error rates compared to traditional dPCR, indicating that amplification errors can be reliably identified and accounted for (164). In addition, U-dHRM is the only test that provides absolute load quantification to enable the resolution of polymicrobial infections and contamination. Further investigation with clinical samples will determine how this approach compares with commercially available technologies.

(iii) Turnaround times.The ideal turnaround times for all the technologies reviewed here are summarized in Fig. 4. The advertised turnaround times of 6 to 8 h for commercially available technologies, including Iridica, SeptiFast, and SepsiTest, may be optimistic for noncentralized and low-resources settings, where the sample-to-result time can be up to 16 h (134, 135). This increased time represents the time for batchwise analysis, sample transfer, and availability of staff. With the possibility of integration as near-point-of-care diagnostic tests, the estimates of <4-h turnaround times for MinION and U-dHRM are more realistic. Here, it is also worth briefly mentioning loop-mediated isothermal amplification (LAMP) technologies, as they offer high portability and short turnaround times. Importantly, however, they cannot be expanded to include a broad range of organisms without losses in sensitivity and specificity (183). The use of multiple specific primers does not allow multiplexing for a large number of pathogens. Alternatively, parallelizing tests using a small blood volume is also undesirable, as it results in a loss of sensitivity for the detection of low-level pathogen loads often associated with sepsis. That being said, LAMP-based tests may be suitable for applications where the detection of a single organism or class of pathogens is desired. The features of isothermal amplification and low technical complexity are particularly advantageous for low-resources settings (184, 185) and could support diagnosis for specific clinical presentations such as tuberculosis or malaria. Likewise, biosurveillance testing for specific viruses could also benefit from the LAMP approach. However, this method is unlikely to replace blood culture in clinics for general infection screening.

FIG 4
  • Open in new tab
  • Download powerpoint
FIG 4

Timeline of sepsis technologies and where they fall compared to the gold standard of blood culture and the 1- to 3-h critical time for affecting clinical decision-making. SeptiCyte and U-dHRM may be further optimized to provide results in a shorter time frame.

Host-Targeted Technologies

SeptiCyte Lab.SeptiCyte Lab (Immunexpress Inc., Seattle, WA) is the first RNA-based technology that targets specific human inflammatory markers using 2.5 ml whole blood for sepsis determination in 4 to 6 h. It has 510(k) clearance from the U.S. FDA for use as an aid in differentiating infection-positive (sepsis) from infection-negative systemic inflammation (SIRS) in critically ill patients on their first day of ICU admission.

SeptiCyte Lab is a host response-targeted, reverse transcription-quantitative PCR (RT-qPCR)-based test that quantifies the relative expression levels of four RNA biomarkers (CEACAM4, LAMP1, PLA2G7, and PLAC8) known to be involved in innate immunity and the host response to infection. In the discovery phase, microarray analysis was used to identify RNA biomarkers that could differentiate patients with sepsis from patients with postsurgical infection-negative systemic inflammation (186). These biomarkers were then converted to a RT-qPCR format and used to develop the SeptiCyte Lab test for sepsis (186). SeptiCyte Lab is rapid, robust, and accurate for classifying patients with infection-related sepsis across gender, race, age, and date of ICU admission (186). It has been suggested to be an indicator of the probability and not the severity of sepsis (187, 188). In a pilot study using 2.5 ml of blood, SeptiCyte Lab effectively discriminated between two groups of critically ill pediatric patients (40 children with clinical severe sepsis syndrome versus 30 children with congenital heart disease). The area under the curve (AUC) in receiver operating characteristic (ROC) curve analysis, which describes the probability that a test will rank a positive incident higher than a negative one when chosen at random, was used to discriminate between the two cohorts. Even for different RNA analysis techniques, an AUC value of >0.9 was obtained (0.99 versus 0.95), indicating high accuracy. In another prospective observational study with 129 adult ICU patients, an AUC of 0.88 was obtained to discriminate SIRS from sepsis. SeptiCyte Lab scores have shown the ability to classify sepsis better than individual or combinations of other clinical, demographic, and laboratory markers (189).

SeptiCyte is a promising, novel, broad-based diagnostic test for sepsis. The current 4- to 6-h turnaround time can potentially be reduced to a targeted 1.5 h by optimizing the RT-qPCR platform on which the test is implemented. One drawback is the requirement for 2.5 ml of blood, which is not feasible for use on neonatal populations. Additionally, this test does not provide any information about the pathogen or its antibiotic resistance. However, a determination of infection-negative SIRS could serve to limit antibiotic treatment. More clinical studies across different patient populations are needed to confirm the ability of SeptiCyte to improve outcomes in the clinic. Nonetheless, it has the potential to play a role in reducing inappropriate empirical antibiotic use, which could be of tremendous value in light of the recent antibiotic resistance epidemic. This technology has not yet commercially launched, and no cost information is available at this time. Combining SeptiCyte with pathogen- and resistance-targeted tests that work within the same critical time frame could generate significant synergy, with the potential to enhance the overall NPV of these diagnostic tests and their impact on antibiotic use. Furthermore, such combined approaches may deepen our understanding of the progression of infection-related sepsis.

Amplification-Free Technology

Droplet digital detection technology.An emerging technology termed “integrated comprehensive droplet digital detection” (IC 3D) (Velox Biosystems, Irvine, CA) claims to selectively detect individual bacterial species directly from small quantities of whole blood within 1 to 4 h (190). In a one-step, culture- and amplification-free process, the IC 3D method provides quantitative bacterial detection with single-cell sensitivity.

IC 3D combines DNAzyme-based sensors with real-time droplet microencapsulation and a particle counter. First, blood samples are directly partitioned into billions of micrometer-sized droplets containing bacteria and a solution containing a fluorescent DNA sensor. The sensor is a DNA probe conjugated to a fluorescent reporter. Upon hybridization to the target sequence, the probe is cleaved and generates a fluorescent signal. Thus, droplets containing bacterial genetic material can be identified by fluorescence. A three-dimensional particle counter is then used to rapidly, robustly, and accurately quantify the fluorescent droplets containing bacteria (191, 192). The distribution of the blood sample into many small droplets minimizes interference from components of blood, making it possible to directly detect target bacteria without sample preparation and purification. In a proof-of-concept study, where blood was infused with Escherichia coli, the IC 3D method confirmed the presence or absence of this target bacterium within an hour. Quantitative measurement of the amount of E. coli bacteria was accomplished in about 3.5 h. In samples containing 1 cell per ml, the assay detected bacteria about 77% of the time (190).

This technology accomplishes rapid pathogen detection with a small blood volume with single-cell sensitivity in a relatively easy-to-use format. Additional probes could be added to detect antibiotic resistance markers. However, the current system design is limited by its ability to detect only one bacterial species (e.g., E. coli) per analysis. There is the potential to expand the sensor set and develop a multiple-wavelength detection system for the detection of multiple bacteria or pathogens (190). However, the extent of this expansion would be limited by the small number of fluorescent channels and would not be able to incorporate the detection of emerging pathogens. Furthermore, the specificity of this technology has not yet been determined, and this technique has not yet been validated by using clinical samples. This technology is currently under commercial development. No cost information is available.

Beyond Rule-Based Decision Support: Power of Electronic Medical Records and Machine Learning-Based AlgorithmsTypically, clinical decision support systems use limited data from patient history to generate severity scores and early warning signs using rule-based algorithms (194–197). Recent advances in machine learning further enable improvements to the timeliness and predictive accuracy of these computerized clinical decision rules. The application of powerful algorithms to large clinical data sets enables these technologies to learn site-specific contexts and novel relationships. As it increasingly becomes clear that a single biomarker may not have both the sensitivity and specificity to inform treatment (198), several groups have proposed algorithms that combine optimal subsets of biomarkers with rich electronic medical record (EMR) data (199–201). For example, a novel targeted real-time early warning score (TREWScore) predicts the development of septic shock in adult ICU patients 28 h before clinical onset (202), providing a superior alternative to the modified early warning score (MEWS) (203). This algorithm uses supervised machine learning to incorporate continuous sampling of a variety of physiological inputs, including platelets, ratio of blood urea nitrogen (BUN) to creatinine, arterial pH, temperature, bicarbonate, respiratory rate (RR), white blood cell count, systolic blood pressure (SBP), heart rate, and heart rate/SBP ratio (shock index). The TREWScore was developed by training the model with data from 13,000 patients to achieve an AUC of 0.83 with a sensitivity of 0.84 and a specificity of 0.67 on a validation set with 3,000 patients. Another recent study used machine learning to identify five optimal biomarkers for use with EMR data and reported an AUC of 0.81 with 10-fold cross-validation on 444 hospitalized adults. This study also attempted to stratify the sepsis syndrome to advance the fundamental understanding of the progression of this disease (199). Several other groups have demonstrated the validity of such algorithms in retrospective studies (204–206). However, only a few machine learning-based algorithms have been implemented in prospective studies beyond the development phase using retrospective chart review. Given this, it is difficult to discuss EMR-based machine learning algorithms in the light of the characteristics of ideal sepsis diagnostic tests. Nonetheless, we think that it is worth summarizing two of these promising approaches, which are under commercial development.

HeRO score.Heart rate characteristics (HRCs) have been used in clinics to provide an “early warning” of patient distress. Available commercially as the HeRO score algorithm (Medical Predictive Science Corporation, Charlottesville, VA), this technology uses signal processing and machine learning to identify subtle irregularities in heart rate variability. The HRC index used by HeRO was shown to reduce the mortality rate from 10% to 8% in an industry-sponsored randomized controlled clinical trial of 3,003 VLBW infants (207). However, the mechanisms for mortality reduction remain unclear. An independent, academic study of HRC monitoring in VLBW infants reported a greater utilization of antibiotics and more sepsis evaluations in a cohort with HRC monitoring than in controls without monitoring. This study also determined that there were no differences in the rates of blood culture-positive sepsis or clinically suspected sepsis as a function of the HeRO index (208). An additional single-center retrospective study reported that elevated HRC scores had a limited ability to detect bloodstream infection among neonates in the NICU, emphasizing that HRCs alone may not be adequate (209). Thus, HeRO may represent another technology that could provide synergy in an integrated format with other diagnostic measures. The incremental costs of HRC monitoring are estimated to be US$2,000 per bed per year, or US$333 per patient, with an average length of stay for VLBW neonates (K. L. Karvonen, J. Vergales, R. A. Sinkin, and R. J. Swanson, presented at the Pediatric Academic Societies [PAS] Meeting, San Francisco, CA, 6 to 9 May 2017).

InSight.InSight (Dascena, Hayward, CA) uses routinely available clinical data to predict likelihood scores for user-specified outcomes. This algorithm has been validated for the early prediction of sepsis, severe sepsis, and septic shock in pediatric and adult populations across multiple centers (210–213). Conveniently, this algorithm requires only electronic health record data routinely available in clinics, with no need for continuous waveform data and extensive laboratory tests. This algorithm allows adaptation to site-specific data by training the model with baseline data from the proposed implementation center. It also allows modifications of gold-standard measures to predict patient conditions, such as in-hospital mortality (212) or patient stability (214). A recent trial across two surgical ICUs with 142 patients (75 controls) reported a 2.7-day reduction in the length of stay (P = 0.04) and a 12% reduction in in-hospital mortality (P = 0.02). Another study, reporting a 4-month experience in a 242-bed acute-care hospital, demonstrated a reduction in the length of stay of 0.43 days, along with a reduction in the mortality rate by 60.24% and a reduction in the rate of sepsis-related 30-day readmission, by 50%, postimplementation (215). Across these two studies, the algorithm score had a sensitivity and a specificity of 0.83 and 0.96, respectively, for sepsis. For severe sepsis, a sensitivity of 0.85 to 0.9 and a specificity of 0.9 were observed for adults. This algorithm has also been validated in pediatric patients (n = 11,000), reporting an AUC of 0.72 for predicting sepsis 4 h before onset and an AUC of 0.92 for predicting sepsis at onset (213). These results show promise for the early detection of sepsis in adult and pediatric patients. However, it remains to be seen if such methods will be equally successful for the neonatal population, where the physiology and pathogenesis of sepsis may be slightly different. For an individual ICU with 50 beds, InSight is predicted to annually save 75 additional lives and reduce sepsis-related costs by US$560,000 (216).

Machine learning techniques allow for the use of heterogeneous data sets to inform clinical decisions. The future should see the incorporation of EMR data with broad-based molecular detection technologies and clinical context to provide a significant increase in the reliability of these technologies. In this era of large-scale data integration, combining broad-based molecular techniques with EMR data represents tremendous opportunities for the timely and accurate diagnosis and management of sepsis as well as gains in insight into human disease mechanisms.

CONCLUSION

An exciting new era of molecular diagnostics for bloodstream infections is emerging through innovations in sample preparation, single-molecule detection methods, sequencing, and applications of machine learning. However, each emerging technology harbors unique benefits and drawbacks. For example, U-dHRM addresses the challenge of detecting pathogens in a low blood volume with high sensitivity while resolving polymicrobial infections, all in a potentially portable format and clinically actionable time frame. However, sample preparation and handling are still required, which increases the time to detection and may lead to some loss of sensitivity. Likewise, SeptiCyte provides a robust way to detect whether a pathogen is present based on the host response and provides this information in a time frame similar to that of U-dHRM but requires a higher volume of blood and initial sample preparation. The IC 3D technology is limited in the number of targets that it can detect in a single sample but is capable of skipping sample preparation entirely to accomplish the simplest and most direct testing from blood samples. This may be of significant value for rapidly tracking the spread of individual organisms in the context of outbreaks and hospital-acquired infections. Furthermore, in the era of big data, advances in the field of machine learning can add patient-specific contextual information to each diagnostic test to potentially increase its sensitivity. The integration of host- and pathogen-targeted diagnostic technologies and their combination with EMR data sets using machine learning constitute a promising new frontier. Combining diagnostic technologies that build on distinct approaches could be a rapid way to improve positive and negative predictive power and truly impact antibiotic usage in the clinic. Together, these emerging technologies have the potential to identify microorganisms and provide relevant subspecies and antibiotic resistance information in a clinically relevant time frame that is much shorter than that currently required for blood culture. Such an integrated approach may overcome the limitations of each technology individually to facilitate targeted and precise antibiotic use.

ACKNOWLEDGMENTS

This research was supported by a UCSD Clinical and Translational Research Institute pilot grant and an Accelerating Innovations to Market award. S.I.F. is supported by a Burroughs Wellcome Fund Career Award at the Scientific Interface and an NSF CAREER award; other laboratory members are supported by the UCSD Frontiers of Innovation Scholars Program.

We are inventors of the U-dHRM device, which is patent pending.

  • Copyright © 2018 American Society for Microbiology.

All Rights Reserved.

REFERENCES

  1. 1.↵
    1. Weinstein MP,
    2. Murphy JR,
    3. Reller LB,
    4. Lichtenstein KA
    . 1983. The clinical significance of positive blood cultures: a comprehensive analysis of 500 episodes of bacteremia and fungemia in adults. II. Clinical observations, with special reference to factors influencing prognosis. Rev Infect Dis 5:54–70. doi:10.1093/clinids/5.1.54.
    OpenUrlCrossRefPubMedWeb of Science
  2. 2.↵
    1. Lee C-C,
    2. Chen S-Y,
    3. Chang I-J,
    4. Chen S-C,
    5. Wu S-C
    . 2007. Comparison of clinical manifestations and outcome of community-acquired bloodstream infections among the oldest old, elderly, and adult patients. Medicine (Baltimore) 86:138–144. doi:10.1097/MD.0b013e31806a754c.
    OpenUrlCrossRefPubMedWeb of Science
  3. 3.↵
    1. Weinstein MP,
    2. Towns ML,
    3. Quartey SM,
    4. Mirrett S,
    5. Reimer LG,
    6. Parmigiani G,
    7. Reller LB
    . 1997. The clinical significance of positive blood cultures in the 1990s: a prospective comprehensive evaluation of the microbiology, epidemiology, and outcome of bacteremia and fungemia in adults. Clin Infect Dis 24:584–602. doi:10.1093/clind/24.4.584.
    OpenUrlCrossRefPubMedWeb of Science
  4. 4.↵
    1. Elixhauser A,
    2. Friedman B,
    3. Stranges E
    . 2011. Septicemia in US hospitals, 2009. Agency for Healthcare Research and Quality, Rockville, MD.
  5. 5.↵
    1. Buehler SS,
    2. Madison B,
    3. Snyder SR,
    4. Derzon JH,
    5. Cornish NE,
    6. Saubolle MA,
    7. Weissfeld AS,
    8. Weinstein MP,
    9. Liebow EB,
    10. Wolk DM
    . 2016. Effectiveness of practices to increase timeliness of providing targeted therapy for inpatients with bloodstream infections: a laboratory medicine best practices systematic review and meta-analysis. Clin Microbiol Rev 29:59–103. doi:10.1128/CMR.00053-14.
    OpenUrlCrossRefPubMed
  6. 6.↵
    1. Novosad SA,
    2. Sapiano MRP,
    3. Grigg C,
    4. Lake J,
    5. Robyn M,
    6. Dumyati G,
    7. Felsen C,
    8. Blog D,
    9. Dufort E,
    10. Zansky S,
    11. Wiedeman K,
    12. Avery L,
    13. Dantes RB,
    14. Jernigan JA,
    15. Magill SS,
    16. Fiore A,
    17. Epstein L
    . 2016. Vital signs. Epidemiology of sepsis: prevalence of health care factors and opportunities for prevention. MMWR Morb Mortal Wkly Rep 65:864–869. doi:10.15585/mmwr.mm6533e1.
    OpenUrlCrossRefPubMed
  7. 7.↵
    1. Torio CM,
    2. Moore BJ
    . 2016. National inpatient hospital costs: the most expensive conditions by pay, 2011. Agency for Healthcare Research and Quality, Rockville, MD.
  8. 8.↵
    1. Epstein L,
    2. Dantes R,
    3. Magill S,
    4. Fiore A
    . 2016. Varying estimates of sepsis mortality using death certificates and administrative codes—United States, 1999-2014. MMWR Morb Mortal Wkly Rep 65:342–345. doi:10.15585/mmwr.mm6513a2.
    OpenUrlCrossRef
  9. 9.↵
    1. Klevens RM,
    2. Edwards JR,
    3. Gaynes RP
    . 2008. The impact of antimicrobial-resistant, health care-associated infections on mortality in the United States. Clin Infect Dis 47:927–930. doi:10.1086/591698.
    OpenUrlCrossRefPubMedWeb of Science
  10. 10.↵
    1. Coburn B,
    2. Morris AM,
    3. Tomlinson G,
    4. Detsky AS
    . 2012. Does this adult patient with suspected bacteremia require blood cultures? JAMA 308:502–511. doi:10.1001/jama.2012.8262.
    OpenUrlCrossRefPubMedWeb of Science
  11. 11.↵
    1. Kiser C,
    2. Nawab U,
    3. McKenna K,
    4. Aghai ZH
    . 2014. Role of guidelines on length of therapy in chorioamnionitis and neonatal sepsis. Pediatrics 133:992–998. doi:10.1542/peds.2013-2927.
    OpenUrlAbstract/FREE Full Text
  12. 12.↵
    1. Patel SJ,
    2. Saiman L
    . 2012. Principles and strategies of antimicrobial stewardship in the neonatal intensive care unit. Semin Perinatol 36:431–436. doi:10.1053/j.semperi.2012.06.005.
    OpenUrlCrossRefPubMed
  13. 13.↵
    1. Heron M
    . 2013. Deaths: leading causes for 2010. Natl Vital Stat Rep 62(6):1–97.
    OpenUrlPubMed
  14. 14.↵
    1. Romero R,
    2. Dey SK,
    3. Fisher SJ
    . 2014. Preterm labor: one syndrome, many causes. Science 345:760–765. doi:10.1126/science.1251816.
    OpenUrlAbstract/FREE Full Text
  15. 15.↵
    1. Kemp MW
    . 2014. Preterm birth, intrauterine infection, and fetal inflammation. Front Immunol 5:574. doi:10.3389/fimmu.2014.00574.
    OpenUrlCrossRefPubMed
  16. 16.↵
    1. Payne MS,
    2. Bayatibojakhi S
    . 2014. Exploring preterm birth as a polymicrobial disease: an overview of the uterine microbiome. Front Immunol 5:595. doi:10.3389/fimmu.2014.00595.
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. Storro O,
    2. Avershina E,
    3. Rudi K
    . 2013. Diversity of intestinal microbiota in infancy and the risk of allergic disease in childhood. Curr Opin Allergy Clin Immunol 13:257–262. doi:10.1097/ACI.0b013e328360968b.
    OpenUrlCrossRef
  18. 18.↵
    1. Saari A,
    2. Virta LJ,
    3. Sankilampi U,
    4. Dunkel L,
    5. Saxen H
    . 2015. Antibiotic exposure in infancy and risk of being overweight in the first 24 months of life. Pediatrics 135:617–626. doi:10.1542/peds.2014-3407.
    OpenUrlAbstract/FREE Full Text
  19. 19.↵
    1. Tripathi N,
    2. Cotten CM,
    3. Smith PB
    . 2012. Antibiotic use and misuse in the neonatal intensive care unit. Clin Perinatol 39:61–68. doi:10.1016/j.clp.2011.12.003.
    OpenUrlCrossRefPubMed
  20. 20.↵
    1. Patel SJ,
    2. Oshodi A,
    3. Prasad P,
    4. Delamora P,
    5. Larson E,
    6. Zaoutis T,
    7. Paul DA,
    8. Saiman L
    . 2009. Antibiotic use in neonatal intensive care units and adherence with Centers for Disease Control and Prevention 12 Step Campaign To Prevent Antimicrobial Resistance. Pediatr Infect Dis J 28:1047–1051. doi:10.1097/INF.0b013e3181b12484.
    OpenUrlCrossRefPubMedWeb of Science
  21. 21.↵
    1. Verani JR,
    2. McGee L,
    3. Schrag SJ
    . 2010. Prevention of perinatal group B streptococcal disease—revised guidelines from CDC, 2010. MMWR Recommend Rep 59:1–36.
    OpenUrl
  22. 22.↵
    1. Fauci AS,
    2. Marston HD
    . 2014. The perpetual challenge of antimicrobial resistance. JAMA 311:1853–1854. doi:10.1001/jama.2014.2465.
    OpenUrlCrossRefPubMedWeb of Science
  23. 23.↵
    1. Ventola CL
    . 2015. The antibiotic resistance crisis. Part 1: causes and threats. P T 40:277–283.
    OpenUrlPubMed
  24. 24.↵
    1. Greenwood C,
    2. Morrow AL,
    3. Lagomarcino AJ,
    4. Altaye M,
    5. Taft DH,
    6. Yu Z,
    7. Newburg DS,
    8. Ward DV,
    9. Schibler KR
    . 2014. Early empiric antibiotic use in preterm infants is associated with lower bacterial diversity and higher relative abundance of Enterobacter. J Pediatr 165:23–29. doi:10.1016/j.jpeds.2014.01.010.
    OpenUrlCrossRefPubMedWeb of Science
  25. 25.↵
    1. Bates DW,
    2. Cook EF,
    3. Goldman L,
    4. Lee TH
    . 1990. Predicting bacteremia in hospitalized patients. A prospectively validated model. Ann Intern Med 113:495–500. doi:10.7326/0003-4819-113-7-495.
    OpenUrlCrossRefPubMedWeb of Science
  26. 26.↵
    1. Roth A,
    2. Wiklund AE,
    3. Pålsson AS,
    4. Melander EZ,
    5. Wullt M,
    6. Cronqvist J,
    7. Walder M,
    8. Sturegård E
    . 2010. Reducing blood culture contamination by a simple informational intervention. J Clin Microbiol 48:4552–4558. doi:10.1128/JCM.00877-10.
    OpenUrlAbstract/FREE Full Text
  27. 27.↵
    1. Stryjewski ME,
    2. Kanafani ZA,
    3. Chu VH,
    4. Pappas PA,
    5. Harding T,
    6. Drew LA,
    7. Benjamin DK,
    8. Reller LB,
    9. Lee BA,
    10. Corey GR,
    11. Fowler VG
    . 2009. Staphylococcus aureus bacteremia among patients with health care-associated fever. Am J Med 122:281.e2–289.e2. doi:10.1016/j.amjmed.2008.09.040.
    OpenUrlCrossRef
  28. 28.↵
    1. Riedel S,
    2. Carroll KC
    . 2016. Early identification and treatment of pathogens in sepsis. Clin Chest Med 37:191–207. doi:10.1016/j.ccm.2016.01.018.
    OpenUrlCrossRef
  29. 29.↵
    1. Riedel S,
    2. Carroll KC
    . 2013. Laboratory detection of sepsis. Clin Lab Med 33:413–437. doi:10.1016/j.cll.2013.03.006.
    OpenUrlCrossRefPubMed
  30. 30.↵
    1. Blackburn RM,
    2. Muller-Pebody B,
    3. Planche T,
    4. Johnson A,
    5. Hopkins S,
    6. Sharland M,
    7. Kennea N,
    8. Heath PT
    . 2012. Neonatal sepsis—many blood samples, few positive cultures: implications for improving antibiotic prescribing. Arch Dis Child Fetal Neonatal Ed 97:F487–F488. doi:10.1136/fetalneonatal-2012-301883.
    OpenUrlFREE Full Text
  31. 31.↵
    1. Ottolini MC,
    2. Lundgren K,
    3. Mirkinson LJ,
    4. Cason S,
    5. Ottolini MG
    . 2003. Utility of complete blood count and blood culture screening to diagnose neonatal sepsis in the asymptomatic at risk newborn. Pediatr Infect Dis J 22:430–434. doi:10.1097/00006454-200305000-00008.
    OpenUrlCrossRefPubMedWeb of Science
  32. 32.↵
    1. Weston EJ,
    2. Pondo T,
    3. Lewis MM,
    4. Martell-Cleary P,
    5. Morin C,
    6. Jewell B,
    7. Daily P,
    8. Apostol M,
    9. Petit S,
    10. Farley M,
    11. Lynfield R,
    12. Reingold A,
    13. Hansen NI,
    14. Stoll BJ,
    15. Shane AL,
    16. Zell E,
    17. Schrag SJ
    . 2011. The burden of invasive early-onset neonatal sepsis in the United States, 2005-2008. Pediatr Infect Dis J 30:937–941. doi:10.1097/INF.0b013e318223bad2.
    OpenUrlCrossRefPubMed
  33. 33.↵
    1. Edmond K,
    2. Zaidi A
    . 2010. New approaches to preventing, diagnosing, and treating neonatal sepsis. PLoS Med 7:e1000213. doi:10.1371/journal.pmed.1000213.
    OpenUrlCrossRefPubMedWeb of Science
  34. 34.↵
    1. Qazi SA,
    2. Stoll BJ
    . 2009. Neonatal sepsis: a major global public health challenge. Pediatr Infect Dis J 28:S1–S2. doi:10.1097/INF.0b013e31819587a9.
    OpenUrlCrossRefPubMed
  35. 35.↵
    1. Dellinger RP,
    2. Levy MM,
    3. Carlet JM,
    4. Bion J,
    5. Parker MM,
    6. Jaeschke R,
    7. Reinhart K,
    8. Angus DC,
    9. Brun-Buisson C,
    10. Beale R,
    11. Calandra T,
    12. Dhainaut J-F,
    13. Gerlach H,
    14. Harvey M,
    15. Marini JJ,
    16. Marshall J,
    17. Ranieri M,
    18. Ramsay G,
    19. Sevransky J,
    20. Thompson BT,
    21. Townsend S,
    22. Vender JS,
    23. Zimmerman JL,
    24. Vincent J-L
    . 2008. Surviving Sepsis campaign. International guidelines for management of severe sepsis and septic shock: 2008. Intensive Care Med 34:17–60. doi:10.1007/s00134-007-0934-2.
    OpenUrlCrossRefPubMedWeb of Science
  36. 36.↵
    1. Kollef MH,
    2. Sherman G,
    3. Ward S,
    4. Fraser VJ
    . 1999. Inadequate antimicrobial treatment of infections: a risk factor for hospital mortality among critically ill patients. Chest 115:462–474. doi:10.1378/chest.115.2.462.
    OpenUrlCrossRefPubMedWeb of Science
  37. 37.↵
    1. Garnacho-Montero J,
    2. Garcia-Garmendia JL,
    3. Barrero-Almodovar A,
    4. Jimenez-Jimenez FJ,
    5. Perez-Paredes C,
    6. Ortiz-Leyba C
    . 2003. Impact of adequate empirical antibiotic therapy on the outcome of patients admitted to the intensive care unit with sepsis. Crit Care Med 31:2742–2751. doi:10.1097/01.CCM.0000098031.24329.10.
    OpenUrlCrossRefPubMedWeb of Science
  38. 38.↵
    1. Valles J,
    2. Rello J,
    3. Ochagavia A,
    4. Garnacho J,
    5. Alcala MA
    . 2003. Community-acquired bloodstream infection in critically ill adult patients: impact of shock and inappropriate antibiotic therapy on survival. Chest 123:1615–1624. doi:10.1378/chest.123.5.1615.
    OpenUrlCrossRefPubMedWeb of Science
  39. 39.↵
    1. Weiss SL,
    2. Fitzgerald JC,
    3. Balamuth F,
    4. Alpern ER,
    5. Lavelle J,
    6. Chilutti M,
    7. Grundmeier R,
    8. Nadkarni VM,
    9. Thomas NJ
    . 2014. Delayed antimicrobial therapy increases mortality and organ dysfunction duration in pediatric sepsis. Crit Care Med 42:2409–2417. doi:10.1097/CCM.0000000000000509.
    OpenUrlCrossRefPubMed
  40. 40.↵
    1. Kumar A,
    2. Ellis P,
    3. Arabi Y,
    4. Roberts D,
    5. Light B,
    6. Parrillo JE,
    7. Dodek P,
    8. Wood G,
    9. Kumar A,
    10. Simon D,
    11. Peters C,
    12. Ahsan M,
    13. Chateau D
    . 2009. Initiation of inappropriate antimicrobial therapy results in a fivefold reduction of survival in human septic shock. Chest 136:1237–1248. doi:10.1378/chest.09-0087.
    OpenUrlCrossRefPubMedWeb of Science
  41. 41.↵
    1. Singer M
    . 2017. Antibiotics for sepsis: does each hour really count, or is it incestuous amplification? Am J Respir Crit Care Med 196:800–802. doi:10.1164/rccm.201703-0621ED.
    OpenUrlCrossRefPubMed
  42. 42.↵
    1. Benitz WE
    . 2010. Adjunct laboratory tests in the diagnosis of early-onset neonatal sepsis. Clin Perinatol 37:421–438. doi:10.1016/j.clp.2009.12.001.
    OpenUrlCrossRefPubMed
  43. 43.↵
    1. Caliendo AM,
    2. Gilbert DN,
    3. Ginocchio CC,
    4. Hanson KE,
    5. May L,
    6. Quinn TC,
    7. Tenover FC,
    8. Alland D,
    9. Blaschke AJ,
    10. Bonomo RA,
    11. Carroll KC,
    12. Ferraro MJ,
    13. Hirschhorn LR,
    14. Joseph WP,
    15. Karchmer T,
    16. MacIntyre AT,
    17. Reller LB,
    18. Jackson AF
    . 2013. Better tests, better care: improved diagnostics for infectious diseases. Clin Infect Dis 57:S139–S170. doi:10.1093/cid/cit578.
    OpenUrlCrossRefPubMed
  44. 44.↵
    1. Chin CD,
    2. Linder V,
    3. Sia SK
    . 2007. Lab-on-a-chip devices for global health: past studies and future opportunities. Lab Chip 7:41–57. doi:10.1039/B611455E.
    OpenUrlCrossRefPubMedWeb of Science
  45. 45.↵
    1. Ilstrup DM,
    2. Washington JA, II
    . 1983. The importance of volume of blood cultured in the detection of bacteremia and fungemia. Diagn Microbiol Infect Dis 1:107–110. doi:10.1016/0732-8893(83)90039-1.
    OpenUrlCrossRefPubMedWeb of Science
  46. 46.↵
    1. Connell TG,
    2. Rele M,
    3. Cowley D,
    4. Buttery JP,
    5. Curtis N
    . 2007. How reliable is a negative blood culture result? Volume of blood submitted for culture in routine practice in a children's hospital. Pediatrics 119:891–896. doi:10.1542/peds.2006-0440.
    OpenUrlAbstract/FREE Full Text
  47. 47.↵
    1. Mermel LA,
    2. Maki DG
    . 1993. Detection of bacteremia in adults: consequences of culturing an inadequate volume of blood. Ann Intern Med 119:270–272. doi:10.7326/0003-4819-119-4-199308150-00003.
    OpenUrlCrossRefPubMedWeb of Science
  48. 48.↵
    1. Bhattacharya S,
    2. Rosenberg AF,
    3. Peterson DR,
    4. Grzesik K,
    5. Baran AM,
    6. Ashton JM,
    7. Gill SR,
    8. Corbett AM,
    9. Holden-Wiltse J,
    10. Topham DJ,
    11. Walsh EE,
    12. Mariani TJ,
    13. Falsey AR
    . 2017. Transcriptomic biomarkers to discriminate bacterial from nonbacterial infection in adults hospitalized with respiratory illness. Sci Rep 7:6548. doi:10.1038/s41598-017-06738-3.
    OpenUrlCrossRef
  49. 49.↵
    1. Ramilo O,
    2. Allman W,
    3. Chung W,
    4. Mejias A,
    5. Ardura M,
    6. Glaser C,
    7. Wittkowski KM,
    8. Piqueras B,
    9. Banchereau J,
    10. Palucka AK,
    11. Chaussabel D
    . 2007. Gene expression patterns in blood leukocytes discriminate patients with acute infections. Blood 109:2066–2077. doi:10.1182/blood-2006-02-002477.
    OpenUrlAbstract/FREE Full Text
  50. 50.↵
    1. Blevins SM,
    2. Bronze MS
    . 2010. Robert Koch and the “golden age” of bacteriology. Int J Infect Dis 14:e744–e751. doi:10.1016/j.ijid.2009.12.003.
    OpenUrlCrossRefPubMed
  51. 51.↵
    1. Petti CA,
    2. Bhally HS,
    3. Weinstein MP,
    4. Joho K,
    5. Wakefield T,
    6. Reller LB,
    7. Carroll KC
    . 2006. Utility of extended blood culture incubation for isolation of Haemophilus, Actinobacillus, Cardiobacterium, Eikenella, and Kingella organisms: a retrospective multicenter evaluation. J Clin Microbiol 44:257–259. doi:10.1128/JCM.44.1.257-259.2006.
    OpenUrlAbstract/FREE Full Text
  52. 52.↵
    1. Baron EJ,
    2. Scott JD,
    3. Tompkins LS
    . 2005. Prolonged incubation and extensive subculturing do not increase recovery of clinically significant microorganisms from standard automated blood cultures. Clin Infect Dis 41:1677–1680. doi:10.1086/497595.
    OpenUrlCrossRefPubMedWeb of Science
  53. 53.↵
    1. Kreger BE,
    2. Craven DE,
    3. Carling PC,
    4. McCabe WR
    . 1980. Gram-negative bacteremia. III. Reassessment of etiology, epidemiology and ecology in 612 patients. Am J Med 68:332–343. doi:10.1016/0002-9343(80)90101-1.
    OpenUrlCrossRefPubMedWeb of Science
  54. 54.↵
    1. Bacconi A,
    2. Richmond GS,
    3. Baroldi MA,
    4. Laffler TG,
    5. Blyn LB,
    6. Carolan HE,
    7. Frinder MR,
    8. Toleno DM,
    9. Metzgar D,
    10. Gutierrez JR,
    11. Massire C,
    12. Rounds M,
    13. Kennel NJ,
    14. Rothman RE,
    15. Peterson S,
    16. Carroll KC,
    17. Wakefield T,
    18. Ecker DJ,
    19. Sampath R
    . 2014. Improved sensitivity for molecular detection of bacterial and candida infections in blood. J Clin Microbiol 52:3164–3174. doi:10.1128/JCM.00801-14.
    OpenUrlAbstract/FREE Full Text
  55. 55.↵
    1. Opota O,
    2. Jaton K,
    3. Greub G
    . 2015. Microbial diagnosis of bloodstream infection: towards molecular diagnosis directly from blood. Clin Microbiol Infect 21:323–331. doi:10.1016/j.cmi.2015.02.005.
    OpenUrlCrossRefPubMed
  56. 56.↵
    1. Cockerill FR, III,
    2. Wilson JW,
    3. Vetter EA,
    4. Goodman KM,
    5. Torgerson CA,
    6. Harmsen WS,
    7. Schleck CD,
    8. Ilstrup DM,
    9. Washington JA, II,
    10. Wilson WR
    . 2004. Optimal testing parameters for blood cultures. Clin Infect Dis 38:1724–1730. doi:10.1086/421087.
    OpenUrlCrossRefPubMedWeb of Science
  57. 57.↵
    1. Riedel S,
    2. Bourbeau P,
    3. Swartz B,
    4. Brecher S,
    5. Carroll KC,
    6. Stamper PD,
    7. Dunne WM,
    8. McCardle T,
    9. Walk N,
    10. Fiebelkorn K,
    11. Sewell D,
    12. Richter SS,
    13. Beekmann S,
    14. Doern GV
    . 2008. Timing of specimen collection for blood cultures from febrile patients with bacteremia. J Clin Microbiol 46:1381–1385. doi:10.1128/JCM.02033-07.
    OpenUrlAbstract/FREE Full Text
  58. 58.↵
    1. Towns ML,
    2. Jarvis WR,
    3. Hsueh P-R
    . 2010. Guidelines on blood cultures. J Microbiol Immunol Infect 43:347–349. doi:10.1016/S1684-1182(10)60054-0.
    OpenUrlCrossRefPubMed
  59. 59.↵
    CLSI. 2007. Principles and procedures for blood cultures; approved guideline. CLSI document M47-A. CLSI, Wayne, PA.
  60. 60.↵
    1. Tille P
    . 2015. Bailey & Scott's diagnostic microbiology, 13th ed. Mosby Elsevier, St. Louis, MO.
  61. 61.↵
    1. Garcia RA,
    2. Spitzer ED,
    3. Beaudry J,
    4. Beck C,
    5. Diblasi R,
    6. Gilleeny-Blabac M,
    7. Haugaard C,
    8. Heuschneider S,
    9. Kranz BP,
    10. McLean K,
    11. Morales KL,
    12. Owens S,
    13. Paciella ME,
    14. Torregrosa E
    . 2015. Multidisciplinary team review of best practices for collection and handling of blood cultures to determine effective interventions for increasing the yield of true-positive bacteremias, reducing contamination, and eliminating false-positive central line-a. Am J Infect Control 43:1222–1237. doi:10.1016/j.ajic.2015.06.030.
    OpenUrlCrossRefPubMed
  62. 62.↵
    1. Kellogg JA,
    2. Ferrentino FL,
    3. Goodstein MH,
    4. Liss J,
    5. Shapiro SL,
    6. Bankert DA
    . 1997. Frequency of low level bacteremia in infants from birth to two months of age. Pediatr Infect Dis J 16:381–385. doi:10.1097/00006454-199704000-00009.
    OpenUrlCrossRefPubMedWeb of Science
  63. 63.↵
    1. Dietzman DE,
    2. Fischer GW,
    3. Schoenknecht FD
    . 1974. Neonatal Escherichia coli septicemia—bacterial counts in blood. J Pediatr 85:128–130. doi:10.1016/S0022-3476(74)80308-2.
    OpenUrlCrossRefPubMedWeb of Science
  64. 64.↵
    1. Fenollar F,
    2. Raoult D
    . 2007. Molecular diagnosis of bloodstream infections caused by non-cultivable bacteria. Int J Antimicrob Agents 30:7–15. doi:10.1016/j.ijantimicag.2007.06.024.
    OpenUrlCrossRef
  65. 65.↵
    1. Riedel S,
    2. Carroll KC
    . 2010. Blood cultures: key elements for best practices and future directions. J Infect Chemother 16:301–316. doi:10.1007/s10156-010-0069-1.
    OpenUrlCrossRefPubMed
  66. 66.↵
    1. Schrag SJ,
    2. Zell ER,
    3. Lynfield R,
    4. Roome A,
    5. Arnold KE,
    6. Craig AS,
    7. Harrison LH,
    8. Reingold A,
    9. Stefonek K,
    10. Smith G,
    11. Gamble M,
    12. Schuchat A
    . 2002. A population-based comparison of strategies to prevent early-onset group B streptococcal disease in neonates. N Engl J Med 347:233–239. doi:10.1056/NEJMoa020205.
    OpenUrlCrossRefPubMedWeb of Science
  67. 67.↵
    1. Stoll BJ,
    2. Hansen NI,
    3. Sánchez PJ,
    4. Faix RG,
    5. Poindexter BB,
    6. Van Meurs KP,
    7. Bizzarro MJ,
    8. Goldberg RN,
    9. Frantz ID,
    10. Hale EC,
    11. Shankaran S,
    12. Kennedy K,
    13. Carlo WA,
    14. Watterberg KL,
    15. Bell EF,
    16. Walsh MC,
    17. Schibler K,
    18. Laptook AR,
    19. Shane AL,
    20. Schrag SJ,
    21. Das A,
    22. Higgins RD
    , Eunice Kennedy Shriver National Institute of Child Health and Human Development Neonatal Research Network. 2011. Early onset neonatal sepsis: the burden of group B streptococcal and E. coli disease continues. Pediatrics 127:817–826. doi:10.1542/peds.2010-2217.
    OpenUrlAbstract/FREE Full Text
  68. 68.↵
    1. Cotten CM,
    2. Taylor S,
    3. Stoll B,
    4. Goldberg RN,
    5. Hansen NI,
    6. Sanchez PJ,
    7. Ambalavanan N,
    8. Benjamin DK, Jr
    . 2009. Prolonged duration of initial empirical antibiotic treatment is associated with increased rates of necrotizing enterocolitis and death for extremely low birth weight infants. Pediatrics 123:58–66. doi:10.1542/peds.2007-3423.
    OpenUrlAbstract/FREE Full Text
  69. 69.↵
    1. Dethlefsen L,
    2. Relman DA
    . 2011. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci U S A 108(Suppl):4554–4561. doi:10.1073/pnas.1000087107.
    OpenUrlAbstract/FREE Full Text
  70. 70.↵
    1. Bekeris LG,
    2. Tworek JA,
    3. Walsh MK,
    4. Valenstein PN
    . 2005. Trends in blood culture contamination: a College of American Pathologists Q-Tracks study of 356 institutions. Arch Pathol Lab Med 129:1222–1225.
    OpenUrlPubMedWeb of Science
  71. 71.↵
    1. Zwang O,
    2. Albert RK
    . 2006. Analysis of strategies to improve cost effectiveness of blood cultures. J Hosp Med 1:272–276. doi:10.1002/jhm.115.
    OpenUrlCrossRefPubMed
  72. 72.↵
    1. Alahmadi YM,
    2. Aldeyab MA,
    3. McElnay JC,
    4. Scott MG,
    5. Darwish Elhajji FW,
    6. Magee FA,
    7. Dowds M,
    8. Edwards C,
    9. Fullerton L,
    10. Tate A,
    11. Kearney MP
    . 2011. Clinical and economic impact of contaminated blood cultures within the hospital setting. J Hosp Infect 77:233–236. doi:10.1016/j.jhin.2010.09.033.
    OpenUrlCrossRefPubMedWeb of Science
  73. 73.↵
    1. Segal GS,
    2. Chamberlain JM
    . 2000. Resource utilization and contaminated blood cultures in children at risk for occult bacteremia. Arch Pediatr Adolesc Med 154:469–473. doi:10.1001/archpedi.154.5.469.
    OpenUrlCrossRefPubMedWeb of Science
  74. 74.↵
    1. Thuler LC,
    2. Jenicek M,
    3. Turgeon JP,
    4. Rivard M,
    5. Lebel P,
    6. Lebel MH
    . 1997. Impact of a false positive blood culture result on the management of febrile children. Pediatr Infect Dis J 16:846–851. doi:10.1097/00006454-199709000-00006.
    OpenUrlCrossRefPubMedWeb of Science
  75. 75.↵
    1. Gander RM,
    2. Byrd L,
    3. DeCrescenzo M,
    4. Hirany S,
    5. Bowen M,
    6. Baughman J
    . 2009. Impact of blood cultures drawn by phlebotomy on contamination rates and health care costs in a hospital emergency department. J Clin Microbiol 47:1021–1024. doi:10.1128/JCM.02162-08.
    OpenUrlAbstract/FREE Full Text
  76. 76.↵
    1. Pien BC,
    2. Sundaram P,
    3. Raoof N,
    4. Costa SF,
    5. Mirrett S,
    6. Woods CW,
    7. Reller LB,
    8. Weinstein MP
    . 2010. The clinical and prognostic importance of positive blood cultures in adults. Am J Med 123:819–828. doi:10.1016/j.amjmed.2010.03.021.
    OpenUrlCrossRefPubMed
  77. 77.↵
    1. Souvenir D,
    2. Anderson DE, Jr,
    3. Palpant S,
    4. Mroch H,
    5. Askin S,
    6. Anderson J,
    7. Claridge J,
    8. Eiland J,
    9. Malone C,
    10. Garrison MW,
    11. Watson P,
    12. Campbell DM
    . 1998. Blood cultures positive for coagulase-negative staphylococci: antisepsis, pseudobacteremia, and therapy of patients. J Clin Microbiol 36:1923–1926.
    OpenUrlAbstract/FREE Full Text
  78. 78.↵
    1. Lee CC,
    2. Lin WJ,
    3. Shih HI,
    4. Wu CJ,
    5. Chen PL,
    6. Lee HC,
    7. Lee NY,
    8. Chang CM,
    9. Wang LR,
    10. Ko WC
    . 2007. Clinical significance of potential contaminants in blood cultures among patients in a medical center. J Microbiol Immunol Infect 40:438–444.
    OpenUrlPubMed
  79. 79.↵
    1. Forrest GN,
    2. Mankes K,
    3. Jabra-Rizk MA,
    4. Weekes E,
    5. Johnson JK,
    6. Lincalis DP,
    7. Venezia RA
    . 2006. Peptide nucleic acid fluorescence in situ hybridization-based identification of Candida albicans and its impact on mortality and antifungal therapy costs. J Clin Microbiol 44:3381–3383. doi:10.1128/JCM.00751-06.
    OpenUrlAbstract/FREE Full Text
  80. 80.↵
    1. Cunney RJ,
    2. McNamara EB,
    3. Alansari N,
    4. Loo B,
    5. Smyth EG
    . 1997. The impact of blood culture reporting and clinical liaison on the empiric treatment of bacteraemia. J Clin Pathol 50:1010–1012. doi:10.1136/jcp.50.12.1010.
    OpenUrlAbstract/FREE Full Text
  81. 81.↵
    1. Zhan C,
    2. Miller MR
    . 2003. Excess length of stay, charges, and mortality attributable to medical injuries during hospitalization. JAMA 290:1868–1874. doi:10.1001/jama.290.14.1868.
    OpenUrlCrossRefPubMedWeb of Science
  82. 82.↵
    1. Dunagan WC,
    2. Woodward RS,
    3. Medoff G,
    4. Gray JL, III,
    5. Casabar E,
    6. Smith MD,
    7. Lawrenz CA,
    8. Spitznagel E
    . 1989. Antimicrobial misuse in patients with positive blood cultures. Am J Med 87:253–259. doi:10.1016/S0002-9343(89)80146-9.
    OpenUrlCrossRefPubMedWeb of Science
  83. 83.↵
    1. Hall KK,
    2. Lyman JA
    . 2006. Updated review of blood culture contamination. Clin Microbiol Rev 19:788–802. doi:10.1128/CMR.00062-05.
    OpenUrlAbstract/FREE Full Text
  84. 84.↵
    1. Lin L,
    2. Nonejuie P,
    3. Munguia J,
    4. Hollands A,
    5. Olson J,
    6. Dam Q,
    7. Kumaraswamy M,
    8. Rivera H,
    9. Corriden R,
    10. Rohde M,
    11. Hensler ME,
    12. Burkart MD,
    13. Pogliano J,
    14. Sakoulas G,
    15. Nizet V
    . 2015. Azithromycin synergizes with cationic antimicrobial peptides to exert bactericidal and therapeutic activity against highly multidrug-resistant Gram-negative bacterial pathogens. EBioMedicine 2:690–698. doi:10.1016/j.ebiom.2015.05.021.
    OpenUrlCrossRefPubMed
  85. 85.↵
    1. Waites KB,
    2. Canupp KC
    . 2001. Evaluation of BacT/ALERT system for detection of Mycoplasma hominis in simulated blood cultures. J Clin Microbiol 39:4328–4331. doi:10.1128/JCM.39.12.4328-4331.2001.
    OpenUrlAbstract/FREE Full Text
  86. 86.↵
    1. Nawrot U,
    2. Kowalska-Krochmal B,
    3. Sulik-Tyszka B,
    4. Kozak M,
    5. Świętek K,
    6. Pajączkowska M,
    7. Piątkowska E,
    8. Rosiak D,
    9. Swoboda-Kopeć E
    . 2015. Evaluation of blood culture media for the detection of fungi. Eur J Clin Microbiol Infect Dis 34:161–167. doi:10.1007/s10096-014-2218-4.
    OpenUrlCrossRefPubMed
  87. 87.↵
    1. Zadroga R,
    2. Williams DN,
    3. Gottschall R,
    4. Hanson K,
    5. Nordberg V,
    6. Deike M,
    7. Kuskowski M,
    8. Carlson L,
    9. Nicolau DP,
    10. Sutherland C,
    11. Hansen GT
    . 2013. Comparison of 2 blood culture media shows significant differences in bacterial recovery for patients on antimicrobial therapy. Clin Infect Dis 56:790–797. doi:10.1093/cid/cis1021.
    OpenUrlCrossRefPubMed
  88. 88.↵
    1. Opota O,
    2. Croxatto A,
    3. Prod'hom G,
    4. Greub G
    . 2015. Blood culture-based diagnosis of bacteraemia: state of the art. Clin Microbiol Infect 21:313–322. doi:10.1016/j.cmi.2015.01.003.
    OpenUrlCrossRefPubMed
  89. 89.↵
    1. Kothari A,
    2. Morgan M,
    3. Haake DA
    . 2014. Emerging technologies for rapid identification of bloodstream pathogens. Clin Infect Dis 59:272–278. doi:10.1093/cid/ciu292.
    OpenUrlCrossRefPubMed
  90. 90.↵
    1. Afshari A,
    2. Schrenzel J,
    3. Ieven M,
    4. Harbarth S
    . 2012. Bench-to-bedside review: rapid molecular diagnostics for bloodstream infection—a new frontier? Crit Care 16:222. doi:10.1186/cc11202.
    OpenUrlCrossRefPubMed
  91. 91.↵
    1. Ecker DJ,
    2. Sampath R,
    3. Li H,
    4. Massire C,
    5. Matthews HE,
    6. Toleno D,
    7. Hall TA,
    8. Blyn LB,
    9. Eshoo MW,
    10. Ranken R,
    11. Hofstadler SA,
    12. Tang YW
    . 2010. New technology for rapid molecular diagnosis of bloodstream infections. Expert Rev Mol Diagn 10:399–415. doi:10.1586/erm.10.24.
    OpenUrlCrossRefPubMed
  92. 92.↵
    1. Jordana-Lluch E,
    2. Giménez M,
    3. Quesada MD,
    4. Rivaya B,
    5. Marcó C,
    6. Domínguez MJ,
    7. Arméstar F,
    8. Martró E,
    9. Ausina V
    . 2015. Evaluation of the broad-range PCR/ESI-MS technology in blood specimens for the molecular diagnosis of bloodstream infections. PLoS One 10:e0140865. doi:10.1371/journal.pone.0140865.
    OpenUrlCrossRefPubMed
  93. 93.↵
    1. Metzgar D,
    2. Frinder MW,
    3. Rothman RE,
    4. Peterson S,
    5. Carroll KC,
    6. Zhang SX,
    7. Avornu GD,
    8. Rounds MA,
    9. Carolan HE,
    10. Toleno DM,
    11. Moore D,
    12. Hall TA,
    13. Massire C,
    14. Richmond GS,
    15. Gutierrez JR,
    16. Sampath R,
    17. Ecker DJ,
    18. Blyn LB
    . 2016. The IRIDICA BAC BSI assay: rapid, sensitive and culture-independent identification of bacteria and candida in blood. PLoS One 11:e0158186. doi:10.1371/journal.pone.0158186.
    OpenUrlCrossRef
  94. 94.↵
    1. Desmet S,
    2. Maertens J,
    3. Bueselinck K,
    4. Lagrou K
    . 2016. Broad-range PCR coupled with electrospray ionization time of flight mass spectrometry for detection of bacteremia and fungemia in patients with neutropenic fever. J Clin Microbiol 54:2513–2520. doi:10.1128/JCM.01066-16.
    OpenUrlAbstract/FREE Full Text
  95. 95.↵
    1. Vincent J-L,
    2. Brealey D,
    3. Libert N,
    4. Abidi NE,
    5. O'Dwyer M,
    6. Zacharowski K,
    7. Mikaszewska-Sokolewicz M,
    8. Schrenzel J,
    9. Simon F,
    10. Wilks M,
    11. Picard-Maureau M,
    12. Chalfin DB,
    13. Ecker DJ,
    14. Sampath R,
    15. Singer M
    . 2015. Rapid diagnosis of infection in the critically ill, a multicenter study of molecular detection in bloodstream infections, pneumonia, and sterile site infections. Crit Care Med 43:2283–2291. doi:10.1097/CCM.0000000000001249.
    OpenUrlCrossRefPubMed
  96. 96.↵
    1. Dien Bard J,
    2. McElvania TeKippe E
    . 2016. Diagnosis of bloodstream infections in children. J Clin Microbiol 54:e02919-15. doi:10.1128/JCM.02919-15.
    OpenUrlAbstract/FREE Full Text
  97. 97.↵
    1. Stevenson M,
    2. Pandor A,
    3. James M-S,
    4. Rafia R,
    5. Uttley L,
    6. Stevens J,
    7. Sanderson J,
    8. Wong R,
    9. Perkins GD,
    10. McMullan R
    . 2016. Sepsis: the LightCycler SeptiFast Test MGRADE, SepsiTest and IRIDICA BAC BSI assay for rapidly identifying bloodstream bacteria and fungi—a systematic review and economic evaluation. Health Technol Assess 20:1–246. doi:10.3310/hta20460.
    OpenUrlCrossRefPubMed
  98. 98.↵
    National Center for Health and Care Excellence. 2016. Tests for rapidly identifying bloodstream bacteria and fungi (LightCycler SeptiFast Test MGRADE, SepsiTest and IRIDICA BAC BSI assay). Guidance and guidelines. National Center for Health and Care Excellence, London, United Kingdom.
  99. 99.↵
    1. Lehmann LE,
    2. Hunfeld K-P,
    3. Emrich T,
    4. Haberhausen G,
    5. Wissing H,
    6. Hoeft A,
    7. Stüber F
    . 2008. A multiplex real-time PCR assay for rapid detection and differentiation of 25 bacterial and fungal pathogens from whole blood samples. Med Microbiol Immunol 197:313–324. doi:10.1007/s00430-007-0063-0.
    OpenUrlCrossRefPubMed
  100. 100.↵
    1. Lucignano B,
    2. Ranno S,
    3. Liesenfeld O,
    4. Pizzorno B,
    5. Putignani L,
    6. Bernaschi P,
    7. Menichella D
    . 2011. Multiplex PCR allows rapid and accurate diagnosis of bloodstream infections in newborns and children with suspected sepsis. J Clin Microbiol 49:2252–2258. doi:10.1128/JCM.02460-10.
    OpenUrlAbstract/FREE Full Text
  101. 101.↵
    1. Dark P,
    2. Blackwood B,
    3. Gates S,
    4. McAuley D,
    5. Perkins GD,
    6. McMullan R,
    7. Wilson C,
    8. Graham D,
    9. Timms K,
    10. Warhurst G
    . 2015. Accuracy of LightCycler SeptiFast for the detection and identification of pathogens in the blood of patients with suspected sepsis: a systematic review and meta-analysis. Intensive Care Med 41:21–33. doi:10.1007/s00134-014-3553-8.
    OpenUrlCrossRefPubMed
  102. 102.↵
    1. Chang S-S,
    2. Hsieh W-H,
    3. Liu T-S,
    4. Lee S-H,
    5. Wang C-H,
    6. Chou H-C,
    7. Yeo YH,
    8. Tseng C-P,
    9. Lee C-C
    . 2013. Multiplex PCR system for rapid detection of pathogens in patients with presumed sepsis—a systemic review and meta-analysis. PLoS One 8:e62323. doi:10.1371/journal.pone.0062323.
    OpenUrlCrossRefPubMed
  103. 103.↵
    1. Louie RF,
    2. Tang Z,
    3. Albertson TE,
    4. Cohen S,
    5. Tran NK,
    6. Kost GJ
    . 2008. Multiplex polymerase chain reaction detection enhancement of bacteremia and fungemia. Crit Care Med 36:1487–1492. doi:10.1097/CCM.0b013e31816f487c.
    OpenUrlCrossRefPubMedWeb of Science
  104. 104.↵
    1. Mancini N,
    2. Clerici D,
    3. Diotti R,
    4. Perotti M,
    5. Ghidoli N,
    6. De Marco D,
    7. Pizzorno B,
    8. Emrich T,
    9. Burioni R,
    10. Ciceri F,
    11. Clementi M
    . 2008. Molecular diagnosis of sepsis in neutropenic patients with haematological malignancies. J Med Microbiol 57:601–604. doi:10.1099/jmm.0.47732-0.
    OpenUrlCrossRefPubMedWeb of Science
  105. 105.↵
    1. Vince A,
    2. Lepej SŽ,
    3. Baršić B,
    4. Dušek D,
    5. Mitrović Z,
    6. Serventi-Seiwerth R,
    7. Labar B
    . 2008. LightCycler SeptiFast assay as a tool for the rapid diagnosis of sepsis in patients during antimicrobial therapy. J Med Microbiol 57:1306–1307. doi:10.1099/jmm.0.47797-0.
    OpenUrlCrossRefPubMed
  106. 106.↵
    1. Dierkes C,
    2. Ehrenstein B,
    3. Siebig S,
    4. Linde H-J,
    5. Reischl U,
    6. Salzberger B
    . 2009. Clinical impact of a commercially available multiplex PCR system for rapid detection of pathogens in patients with presumed sepsis. BMC Infect Dis 9:126. doi:10.1186/1471-2334-9-126.
    OpenUrlCrossRefPubMed
  107. 107.↵
    1. von Lilienfeld-Toal M,
    2. Lehmann LE,
    3. Raadts AD,
    4. Hahn-Ast C,
    5. Orlopp KS,
    6. Marklein G,
    7. Purr I,
    8. Cook G,
    9. Hoeft A,
    10. Glasmacher A,
    11. Stüber F
    . 2009. Utility of a commercially available multiplex real-time PCR assay to detect bacterial and fungal pathogens in febrile neutropenia. J Clin Microbiol 47:2405–2410. doi:10.1128/JCM.00491-09.
    OpenUrlAbstract/FREE Full Text
  108. 108.↵
    1. Varani S,
    2. Stanzani M,
    3. Paolucci M,
    4. Melchionda F,
    5. Castellani G,
    6. Nardi L,
    7. Landini MP,
    8. Baccarani M,
    9. Pession A,
    10. Sambri V
    . 2009. Diagnosis of bloodstream infections in immunocompromised patients by real-time PCR. J Infect 58:346–351. doi:10.1016/j.jinf.2009.03.001.
    OpenUrlCrossRefPubMedWeb of Science
  109. 109.↵
    1. Westh H,
    2. Lisby G,
    3. Breysse F,
    4. Böddinghaus B,
    5. Chomarat M,
    6. Gant V,
    7. Goglio A,
    8. Raglio A,
    9. Schuster H,
    10. Stuber F,
    11. Wissing H,
    12. Hoeft A
    . 2009. Multiplex real-time PCR and blood culture for identification of bloodstream pathogens in patients with suspected sepsis. Clin Microbiol Infect 15:544–551. doi:10.1111/j.1469-0691.2009.02736.x.
    OpenUrlCrossRefPubMedWeb of Science
  110. 110.↵
    1. Avolio M,
    2. Diamante P,
    3. Zamparo S,
    4. Modolo ML,
    5. Grosso S,
    6. Zigante P,
    7. Tosoni N,
    8. De Rosa R,
    9. Stano P,
    10. Camporese A
    . 2010. Molecular identification of bloodstream pathogens in patients presenting to the emergency department with suspected sepsis. Shock 34:27–30. doi:10.1097/SHK.0b013e3181d49299.
    OpenUrlCrossRefPubMed
  111. 111.↵
    1. Bloos F,
    2. Hinder F,
    3. Becker K,
    4. Sachse S,
    5. Dessap AM,
    6. Straube E,
    7. Cattoir V,
    8. Brun-Buisson C,
    9. Reinhart K,
    10. Peters G
    . 2010. A multicenter trial to compare blood culture with polymerase chain reaction in severe human sepsis. Intensive Care Med 36:241–247. doi:10.1007/s00134-009-1705-z.
    OpenUrlCrossRefPubMed
  112. 112.↵
    1. Lehmann LE,
    2. Herpichboehm B,
    3. Kost GJ,
    4. Kollef MH,
    5. Stuber F,
    6. Rodriguez J,
    7. Regueiro B,
    8. Khosrovaneh A,
    9. Reinhart K,
    10. Peters G,
    11. Bauer M,
    12. Hoeft A
    . 2010. Cost and mortality prediction using polymerase chain reaction pathogen detection in sepsis: evidence from three observational trials. Crit Care 14:R186. doi:10.1186/cc9294.
    OpenUrlCrossRefPubMed
  113. 113.↵
    1. Maubon D,
    2. Hamidfar-Roy R,
    3. Courby S,
    4. Vesin A,
    5. Maurin M,
    6. Pavese P,
    7. Ravanel N,
    8. Bulabois C-E,
    9. Brion J-P,
    10. Pelloux H,
    11. Timsit J-F
    . 2010. Therapeutic impact and diagnostic performance of multiplex PCR in patients with malignancies and suspected sepsis. J Infect 61:335–342. doi:10.1016/j.jinf.2010.07.004.
    OpenUrlCrossRefPubMedWeb of Science
  114. 114.↵
    1. Obara H,
    2. Tanabe M,
    3. Kitajima M,
    4. Kitagawa Y,
    5. Kitajima M,
    6. Aikawa N,
    7. Hori S,
    8. Hasegawa N,
    9. Ikeda Y,
    10. Okamoto S,
    11. Kobayashi Y,
    12. Murata M,
    13. Takeda J,
    14. Sakakura Y,
    15. Ginba H
    . 2011. The role of a real-time PCR technology for rapid detection and identification of bacterial and fungal pathogens in whole-blood samples. J Infect Chemother 17:327–333. doi:10.1007/s10156-010-0168-z.
    OpenUrlCrossRefPubMed
  115. 115.↵
    1. Regueiro BJ,
    2. Varela-Ledo E,
    3. Martinez-Lamas L,
    4. Rodriguez-Calviño J,
    5. Aguilera A,
    6. Santos A,
    7. Gomez-Tato A,
    8. Alvarez-Escudero J
    . 2010. Automated extraction improves multiplex molecular detection of infection in septic patients. PLoS One 5:e13387. doi:10.1371/journal.pone.0013387.
    OpenUrlCrossRefPubMed
  116. 116.↵
    1. Tsalik EL,
    2. Jones D,
    3. Nicholson B,
    4. Waring L,
    5. Liesenfeld O,
    6. Park LP,
    7. Glickman SW,
    8. Caram LB,
    9. Langley RJ,
    10. van Velkinburgh JC,
    11. Cairns CB,
    12. Rivers EP,
    13. Otero RM,
    14. Kingsmore SF,
    15. Lalani T,
    16. Fowler VG,
    17. Woods CW
    . 2010. Multiplex PCR to diagnose bloodstream infections in patients admitted from the emergency department with sepsis. J Clin Microbiol 48:26–33. doi:10.1128/JCM.01447-09.
    OpenUrlAbstract/FREE Full Text
  117. 117.↵
    1. Wallet F,
    2. Nseir S,
    3. Baumann L,
    4. Herwegh S,
    5. Sendid B,
    6. Boulo M,
    7. Roussel-Delvallez M,
    8. Durocher AV,
    9. Courcol RJ
    . 2010. Preliminary clinical study using a multiplex real-time PCR test for the detection of bacterial and fungal DNA directly in blood. Clin Microbiol Infect 16:774–779. doi:10.1111/j.1469-0691.2009.02940.x.
    OpenUrlCrossRefPubMed
  118. 118.↵
    1. Yanagihara K,
    2. Kitagawa Y,
    3. Tomonaga M,
    4. Tsukasaki K,
    5. Kohno S,
    6. Seki M,
    7. Sugimoto H,
    8. Shimazu T,
    9. Tasaki O,
    10. Matsushima A
    . 2010. Evaluation of pathogen detection from clinical samples by real-time polymerase chain reaction using a sepsis pathogen DNA detection kit. Crit Care 14:R159. doi:10.1186/cc9234.
    OpenUrlCrossRefPubMed
  119. 119.↵
    1. Bravo D,
    2. Blanquer J,
    3. Tormo M,
    4. Aguilar G,
    5. Borrás R,
    6. Solano C,
    7. Clari MA,
    8. Costa E,
    9. Muñoz-Cobo B,
    10. Argüeso M,
    11. Pineda JR,
    12. Navarro D
    . 2011. Diagnostic accuracy and potential clinical value of the LightCycler SeptiFast assay in the management of bloodstream infections occurring in neutropenic and critically ill patients. Int J Infect Dis 15:e326–e331. doi:10.1016/j.ijid.2011.01.003.
    OpenUrlCrossRefPubMed
  120. 120.↵
    1. Josefson P,
    2. Strålin K,
    3. Ohlin A,
    4. Ennefors T,
    5. Dragsten B,
    6. Andersson L,
    7. Fredlund H,
    8. Mölling P,
    9. Olcén P
    . 2011. Evaluation of a commercial multiplex PCR test (SeptiFast) in the etiological diagnosis of community-onset bloodstream infections. Eur J Clin Microbiol Infect Dis 30:1127–1134. doi:10.1007/s10096-011-1201-6.
    OpenUrlCrossRefPubMed
  121. 121.↵
    1. Lodes U,
    2. Bohmeier B,
    3. Lippert H,
    4. König B,
    5. Meyer F
    . 2012. PCR-based rapid sepsis diagnosis effectively guides clinical treatment in patients with new onset of SIRS. Langenbecks Arch Surg 397:447–455. doi:10.1007/s00423-011-0870-z.
    OpenUrlCrossRefPubMed
  122. 122.↵
    1. Grif K,
    2. Fille M,
    3. Würzner R,
    4. Weiss G,
    5. Lorenz I,
    6. Gruber G,
    7. Eschertzhuber S,
    8. Nachbaur D,
    9. Lass-Flörl C,
    10. Orth D
    . 2012. Rapid detection of bloodstream pathogens by real-time PCR in patients with sepsis. Wien Klin Wochenschr 124:266–270. doi:10.1007/s00508-012-0159-4.
    OpenUrlCrossRefPubMed
  123. 123.↵
    1. Guido M,
    2. Quattrocchi M,
    3. Zizza A,
    4. Pasanisi G,
    5. Pavone V,
    6. Lobreglio G,
    7. Gabutti G,
    8. De Donno MA
    . 2012. Molecular approaches in the diagnosis of sepsis in neutropenic patients with haematological malignances. J Prev Med Hyg 53:104–108.
    OpenUrlPubMed
  124. 124.↵
    1. Hettwer S,
    2. Wilhelm J,
    3. Schürmann M,
    4. Ebelt H,
    5. Hammer D,
    6. Amoury M,
    7. Hofmann F,
    8. Oehme A,
    9. Wilhelms D,
    10. Kekulé AS,
    11. Klöss T,
    12. Werdan K
    . 2012. Microbial diagnostics in patients with presumed severe infection in the emergency department. Med Klin Intensivmed Notfmed 107:53–62. doi:10.1007/s00063-011-0051-4.
    OpenUrlCrossRefPubMed
  125. 125.↵
    1. Mauro MV,
    2. Cavalcanti P,
    3. Perugini D,
    4. Noto A,
    5. Sperlì D,
    6. Giraldi C
    . 2012. Diagnostic utility of LightCycler SeptiFast and procalcitonin assays in the diagnosis of bloodstream infection in immunocompromised patients. Diagn Microbiol Infect Dis 73:308–311. doi:10.1016/j.diagmicrobio.2012.04.006.
    OpenUrlCrossRefPubMed
  126. 126.↵
    1. Pasqualini L,
    2. Mencacci A,
    3. Leli C,
    4. Montagna P,
    5. Cardaccia A,
    6. Cenci E,
    7. Montecarlo I,
    8. Pirro M,
    9. di Filippo F,
    10. Cistaro E,
    11. Schillaci G,
    12. Bistoni F,
    13. Mannarino E
    . 2012. Diagnostic performance of a multiple real-time PCR assay in patients with suspected sepsis hospitalized in an internal medicine ward. J Clin Microbiol 50:1285–1288. doi:10.1128/JCM.06793-11.
    OpenUrlAbstract/FREE Full Text
  127. 127.↵
    1. Rath P-M,
    2. Saner F,
    3. Paul A,
    4. Lehmann N,
    5. Steinmann E,
    6. Buer J,
    7. Steinmann J
    . 2012. Multiplex PCR for rapid and improved diagnosis of bloodstream infections in liver transplant recipients. J Clin Microbiol 50:2069–2071. doi:10.1128/JCM.00745-12.
    OpenUrlAbstract/FREE Full Text
  128. 128.↵
    1. Tschiedel E,
    2. Steinmann J,
    3. Buer J,
    4. Onnebrink J,
    5. Felderhoff-Müser U,
    6. Rath P-M,
    7. Dohna-Schwake C
    . 2012. Results and relevance of molecular detection of pathogens by SeptiFast—a retrospective analysis in 75 critically ill children. Klin Padiatr 224:12–16. doi:10.1055/s-0031-1285878.
    OpenUrlCrossRefPubMed
  129. 129.↵
    1. Herne V,
    2. Nelovkov A,
    3. Kütt M,
    4. Ivanova M
    . 2013. Diagnostic performance and therapeutic impact of LightCycler SeptiFast assay in patients with suspected sepsis. Eur J Microbiol Immunol (Bp) 3:68–76. doi:10.1556/EuJMI.3.2013.1.10.
    OpenUrlCrossRefPubMed
  130. 130.↵
    1. Leitner E,
    2. Kessler HH,
    3. Spindelboeck W,
    4. Hoenigl M,
    5. Putz-Bankuti C,
    6. Stadlbauer-Köllner V,
    7. Krause R,
    8. Grisold AJ,
    9. Feierl G,
    10. Stauber RE
    . 2013. Comparison of two molecular assays with conventional blood culture for diagnosis of sepsis. J Microbiol Methods 92:253–255. doi:10.1016/j.mimet.2012.12.012.
    OpenUrlCrossRefPubMed
  131. 131.↵
    1. Avolio M,
    2. Diamante P,
    3. Modolo ML,
    4. De Rosa R,
    5. Stano P,
    6. Camporese A
    . 2014. Direct molecular detection of pathogens in blood as specific rule-in diagnostic biomarker in patients with presumed sepsis. Shock 42:86–92. doi:10.1097/SHK.0000000000000191.
    OpenUrlCrossRef
  132. 132.↵
    1. Burdino E,
    2. Ruggiero T,
    3. Allice T,
    4. Milia MG,
    5. Gregori G,
    6. Milano R,
    7. Cerutti F,
    8. De Rosa FG,
    9. Manno E,
    10. Caramello P,
    11. Di Perri G,
    12. Ghisetti V
    . 2014. Combination of conventional blood cultures and the SeptiFast molecular test in patients with suspected sepsis for the identification of bloodstream pathogens. Diagn Microbiol Infect Dis 79:287–292. doi:10.1016/j.diagmicrobio.2014.03.018.
    OpenUrlCrossRef
  133. 133.↵
    1. Ortiz Ibarra J,
    2. Trevino Valdez P,
    3. Valenzuela Mendez E,
    4. Limon Rojas A,
    5. Lara Flores G,
    6. Ceballos Bocanegra A,
    7. Morales Mendez I,
    8. Fernandez Carrocera L,
    9. Covian Molina E,
    10. Reyna Figueroa J
    . 2015. Evaluation of the Light-Cycler SeptiFast test in newborns with suspicion of nosocomial sepsis. Iran J Pediatr 25:e253. doi:10.5812/ijp.253.
    OpenUrlCrossRef
  134. 134.↵
    1. Stefani S,
    2. Mongelli G,
    3. Romeo MA,
    4. Denaro C,
    5. Gennaro M,
    6. Fraggetta F
    . 2015. Added value of multi-pathogen probe-based real-time PCR SeptiFast in the rapid diagnosis of bloodstream infections in patients with bacteraemia. J Med Microbiol 64:670–675. doi:10.1099/jmm.0.000074.
    OpenUrlCrossRef
  135. 135.↵
    1. Tafelski S,
    2. Nachtigall I,
    3. Adam T,
    4. Bereswill S,
    5. Faust J,
    6. Tamarkin A,
    7. Trefzer T,
    8. Deja M,
    9. Idelevich EA,
    10. Wernecke K-D,
    11. Becker K,
    12. Spies C
    . 2015. Randomized controlled clinical trial evaluating multiplex polymerase chain reaction for pathogen identification and therapy adaptation in critical care patients with pulmonary or abdominal sepsis. J Int Med Res 43:364–377. doi:10.1177/0300060514561135.
    OpenUrlCrossRefPubMed
  136. 136.↵
    1. Dinç F,
    2. Akalin H,
    3. Özakin C,
    4. Sinirtaş M,
    5. Kebabçi N,
    6. Işçimen R,
    7. Kelebek Girgin N,
    8. Kahveci F
    . 2016. Comparison of blood culture and multiplex real-time PCR for the diagnosis of nosocomial sepsis. Minerva Anestesiol 82:301–309.
    OpenUrl
  137. 137.↵
    1. Ratzinger F,
    2. Tsirkinidou I,
    3. Haslacher H,
    4. Perkmann T,
    5. Schmetterer KG,
    6. Mitteregger D,
    7. Makristathis A,
    8. Burgmann H
    . 2016. Evaluation of the Septifast MGrade test on standard care wards—a cohort study. PLoS One 11:e0151108. doi:10.1371/journal.pone.0151108.
    OpenUrlCrossRef
  138. 138.↵
    1. Suberviola B,
    2. Márquez-López A,
    3. Castellanos-Ortega A,
    4. Fernández-Mazarrasa C,
    5. Santibáñez M,
    6. Martínez LM
    . 2016. Microbiological diagnosis of sepsis: polymerase chain reaction system versus blood cultures. Am J Crit Care 25:68–75. doi:10.4037/ajcc2016728.
    OpenUrlAbstract/FREE Full Text
  139. 139.↵
    1. Tröger B,
    2. Härtel C,
    3. Buer J,
    4. Dördelmann M,
    5. Felderhoff-Müser U,
    6. Höhn T,
    7. Hepping N,
    8. Hillebrand G,
    9. Kribs A,
    10. Marissen J,
    11. Olbertz D,
    12. Rath P-M,
    13. Schmidtke S,
    14. Siegel J,
    15. Herting E,
    16. Göpel W,
    17. Steinmann J,
    18. Stein A
    . 2016. Clinical relevance of pathogens detected by multiplex PCR in blood of very-low-birth weight infants with suspected sepsis—multicentre study of the German Neonatal Network. PLoS One 11:e0159821. doi:10.1371/journal.pone.0159821.
    OpenUrlCrossRef
  140. 140.↵
    1. Korber F,
    2. Zeller I,
    3. Grünstäudl M,
    4. Willinger B,
    5. Apfalter P,
    6. Hirschl AM,
    7. Makristathis A
    . 2017. SeptiFast versus blood culture in clinical routine—a report on 3 years experience. Wien Klin Wochenschr 129:427–434. doi:10.1007/s00508-017-1181-3.
    OpenUrlCrossRef
  141. 141.↵
    1. Gies F,
    2. Tschiedel E,
    3. Felderhoff-Müser U,
    4. Rath P-M,
    5. Steinmann J,
    6. Dohna-Schwake C
    . 2016. Prospective evaluation of SeptiFast multiplex PCR in children with systemic inflammatory response syndrome under antibiotic treatment. BMC Infect Dis 16:378. doi:10.1186/s12879-016-1722-9.
    OpenUrlCrossRef
  142. 142.↵
    1. Reinhart K,
    2. Bauer M,
    3. Riedemann NC,
    4. Hartog CS
    . 2012. New approaches to sepsis: molecular diagnostics and biomarkers. Clin Microbiol Rev 25:609–634. doi:10.1128/CMR.00016-12.
    OpenUrlAbstract/FREE Full Text
  143. 143.↵
    1. Mancini N,
    2. Carletti S,
    3. Ghidoli N,
    4. Cichero P,
    5. Ossi CM,
    6. Ieri R,
    7. Poli E,
    8. Burioni R,
    9. Clementi M
    . 2009. Molecular diagnosis of polymicrobial sepsis. J Clin Microbiol 47:1274–1275. doi:10.1128/JCM.00011-09.
    OpenUrlFREE Full Text
  144. 144.↵
    1. Vrioni G,
    2. Daniil I,
    3. Drogari-Apiranthitou M,
    4. Kimouli M,
    5. Papadopoulou M,
    6. Tsakris A
    . 2012. Molecular diagnosis of polymicrobial newborn sepsis by multiplex real-time PCR using a small volume of blood sample. J Med Microbiol 61:1177–1178. doi:10.1099/jmm.0.037127-0.
    OpenUrlCrossRefPubMed
  145. 145.↵
    1. Fernandez-Cruz A,
    2. Marin M,
    3. Kestler M,
    4. Alcala L,
    5. Rodriguez-Creixems M,
    6. Bouza E
    . 2013. The value of combining blood culture and SeptiFast data for predicting complicated bloodstream infections caused by Gram-positive bacteria or Candida species. J Clin Microbiol 51:1130–1136. doi:10.1128/JCM.02882-12.
    OpenUrlAbstract/FREE Full Text
  146. 146.↵
    1. Warhurst G,
    2. Maddi S,
    3. Dunn G,
    4. Ghrew M,
    5. Chadwick P,
    6. Alexander P,
    7. Bentley A,
    8. Moore J,
    9. Sharman M,
    10. Carlson GL,
    11. Young D,
    12. Dark P
    . 2015. Diagnostic accuracy of SeptiFast multi-pathogen real-time PCR in the setting of suspected healthcare-associated bloodstream infection. Intensive Care Med 41:86–93. doi:10.1007/s00134-014-3551-x.
    OpenUrlCrossRef
  147. 147.↵
    1. Horz H-P,
    2. Scheer S,
    3. Vianna ME,
    4. Conrads G
    . 2010. New methods for selective isolation of bacterial DNA from human clinical specimens. Anaerobe 16:47–53. doi:10.1016/j.anaerobe.2009.04.009.
    OpenUrlCrossRefPubMedWeb of Science
  148. 148.↵
    1. Mühl H,
    2. Kochem A-J,
    3. Disqué C,
    4. Sakka SG
    . 2010. Activity and DNA contamination of commercial polymerase chain reaction reagents for the universal 16S rDNA real-time polymerase chain reaction detection of bacterial pathogens in blood. Diagn Microbiol Infect Dis 66:41–49. doi:10.1016/j.diagmicrobio.2008.07.011.
    OpenUrlCrossRefPubMedWeb of Science
  149. 149.↵
    1. Sakka SG,
    2. Kochem A-J,
    3. Disqué C,
    4. Wellinghausen N
    . 2009. Blood infection diagnosis by 16S rDNA broad-spectrum polymerase chain reaction: the relationship between antibiotic treatment and bacterial DNA load. Anesth Analg 109:1707–1708. doi:10.1213/ANE.0b013e3181b79904.
    OpenUrlCrossRefPubMed
  150. 150.↵
    1. Loonen AJM,
    2. de Jager CPC,
    3. Tosserams J,
    4. Kusters R,
    5. Hilbink M,
    6. Wever PC,
    7. van den Brule AJC
    . 2014. Biomarkers and molecular analysis to improve bloodstream infection diagnostics in an emergency care unit. PLoS One 9:e87315. doi:10.1371/journal.pone.0087315.
    OpenUrlCrossRefPubMed
  151. 151.↵
    1. Kühn C,
    2. Disqué C,
    3. Mühl H,
    4. Orszag P,
    5. Stiesch M,
    6. Haverich A
    . 2011. Evaluation of commercial universal rRNA gene PCR plus sequencing tests for identification of bacteria and fungi associated with infectious endocarditis. J Clin Microbiol 49:2919–2923. doi:10.1128/JCM.00830-11.
    OpenUrlAbstract/FREE Full Text
  152. 152.↵
    1. Wellinghausen N,
    2. Kochem A-J,
    3. Disqué C,
    4. Mühl H,
    5. Gebert S,
    6. Winter J,
    7. Matten J,
    8. Sakka SG
    . 2009. Diagnosis of bacteremia in whole-blood samples by use of a commercial universal 16S rRNA gene-based PCR and sequence analysis. J Clin Microbiol 47:2759–2765. doi:10.1128/JCM.00567-09.
    OpenUrlAbstract/FREE Full Text
  153. 153.↵
    1. Nieman AE,
    2. Savelkoul PHM,
    3. Beishuizen A,
    4. Henrich B,
    5. Lamik B,
    6. MacKenzie CR,
    7. Kindgen-Milles D,
    8. Helmers A,
    9. Diaz C,
    10. Sakka SG,
    11. Schade RP
    . 2016. A prospective multicenter evaluation of direct molecular detection of blood stream infection from a clinical perspective. BMC Infect Dis 16:314. doi:10.1186/s12879-016-1646-4.
    OpenUrlCrossRef
  154. 154.↵
    1. Orszag P,
    2. Disque C,
    3. Keim S,
    4. Lorenz MG,
    5. Wiesner O,
    6. Hadem J,
    7. Stiesch M,
    8. Haverich A,
    9. Kuhn C
    . 2014. Monitoring of patients supported by extracorporeal membrane oxygenation for systemic infections by broad-range rRNA gene PCR amplification and sequence analysis. J Clin Microbiol 52:307–311. doi:10.1128/JCM.02493-13.
    OpenUrlAbstract/FREE Full Text
  155. 155.↵
    1. Wang Y,
    2. Yang Q,
    3. Wang Z
    . 2015. The evolution of nanopore sequencing. Front Genet 5:449. doi:10.3389/fgene.2014.00449.
    OpenUrlCrossRef
  156. 156.↵
    1. Schmidt K,
    2. Mwaigwisya S,
    3. Crossman LC,
    4. Doumith M,
    5. Munroe D,
    6. Pires C,
    7. Khan AM,
    8. Woodford N,
    9. Saunders NJ,
    10. Wain J,
    11. O'Grady J,
    12. Livermore DM
    . 2016. Identification of bacterial pathogens and antimicrobial resistance directly from clinical urines by nanopore-based metagenomic sequencing. J Antimicrob Chemother 72:104–114. doi:10.1093/jac/dkw397.
    OpenUrlCrossRefPubMed
  157. 157.↵
    Oxford Nanopore Technologies. 2017. DNA extraction and library preparation for rapid genus- and species-level identification, with or without PCR. Oxford Nanopore Technologies, Oxford, United Kingdom.
  158. 158.↵
    1. Quick J,
    2. Ashton P,
    3. Calus S,
    4. Chatt C,
    5. Gossain S,
    6. Hawker J,
    7. Nair S,
    8. Neal K,
    9. Nye K,
    10. Peters T,
    11. De Pinna E,
    12. Robinson E,
    13. Struthers K,
    14. Webber M,
    15. Catto A,
    16. Dallman TJ,
    17. Hawkey P,
    18. Loman NJ
    . 2015. Rapid draft sequencing and real-time nanopore sequencing in a hospital outbreak of Salmonella. Genome Biol 16:114. doi:10.1186/s13059-015-0677-2.
    OpenUrlCrossRefPubMed
  159. 159.↵
    1. Benitez-Paez A,
    2. Portune K,
    3. Sanz Y
    . 2016. Species-level resolution of 16S rRNA gene amplicons sequenced through MinION portable nanopore sequencer. Gigascience 5:4. doi:10.1186/s13742-016-0111-z.
    OpenUrlCrossRef
  160. 160.↵
    1. Mitsuhashi S,
    2. Kryukov K,
    3. Nakagawa S,
    4. Takeuchi JS,
    5. Shiraishi Y,
    6. Asano K,
    7. Imanishi T
    . 2017. A portable system for metagenomic analyses using nanopore-based sequencer and laptop computers can realize rapid on-site determination of bacterial compositions. bioRxiv doi:10.1101/101865.
    OpenUrlAbstract/FREE Full Text
  161. 161.↵
    1. Greninger AL,
    2. Naccache SN,
    3. Federman S,
    4. Yu G,
    5. Mbala P,
    6. Bres V,
    7. Stryke D,
    8. Bouquet J,
    9. Somasekar S,
    10. Linnen JM,
    11. Dodd R,
    12. Mulembakani P,
    13. Schneider BS,
    14. Muyembe-Tamfum J-J,
    15. Stramer SL,
    16. Chiu CY
    . 2015. Rapid metagenomic identification of viral pathogens in clinical samples by real-time nanopore sequencing analysis. Genome Med 7:99. doi:10.1186/s13073-015-0220-9.
    OpenUrlCrossRefPubMed
  162. 162.↵
    1. Ashton PM,
    2. Nair S,
    3. Dallman T,
    4. Rubino S,
    5. Rabsch W,
    6. Mwaigwisya S,
    7. Wain J,
    8. O'Grady J
    . 2015. MinION nanopore sequencing identifies the position and structure of a bacterial antibiotic resistance island. Nat Biotechnol 33:296–300. doi:10.1038/nbt.3103.
    OpenUrlCrossRefPubMed
  163. 163.↵
    1. Bradley P,
    2. Gordon NC,
    3. Walker TM,
    4. Dunn L,
    5. Heys S,
    6. Huang B,
    7. Earle S,
    8. Pankhurst LJ,
    9. Anson L,
    10. de Cesare M,
    11. Piazza P,
    12. Votintseva AA,
    13. Golubchik T,
    14. Wilson DJ,
    15. Wyllie DH,
    16. Diel R,
    17. Niemann S,
    18. Feuerriegel S,
    19. Kohl TA,
    20. Ismail N,
    21. Omar SV,
    22. Smith EG,
    23. Buck D,
    24. McVean G,
    25. Walker AS,
    26. Peto TEA,
    27. Crook DW,
    28. Iqbal Z
    . 2015. Rapid antibiotic-resistance predictions from genome sequence data for Staphylococcus aureus and Mycobacterium tuberculosis. Nat Commun 6:10063. doi:10.1038/ncomms10063.
    OpenUrlCrossRefPubMed
  164. 164.↵
    1. Fraley SI,
    2. Hardick J,
    3. Masek BJ,
    4. Athamanolap P,
    5. Rothman RE,
    6. Gaydos CA,
    7. Carroll KC,
    8. Wakefield T,
    9. Wang TH,
    10. Yang S
    . 2013. Universal digital high-resolution melt: a novel approach to broad-based profiling of heterogeneous biological samples. Nucleic Acids Res 41:e175. doi:10.1093/nar/gkt684.
    OpenUrlCrossRefPubMed
  165. 165.↵
    1. Velez DO,
    2. Mack H,
    3. Jupe J,
    4. Hawker S,
    5. Kulkarni N,
    6. Hedayatnia B,
    7. Zhang Y,
    8. Lawrence S,
    9. Fraley SI
    . 2017. Massively parallel digital high resolution melt for rapid and absolutely quantitative sequence profiling. Sci Rep 7:42326. doi:10.1038/srep42326.
    OpenUrlCrossRef
  166. 166.↵
    1. Erali M,
    2. Voelkerding KV,
    3. Wittwer CT
    . 2008. High resolution melting applications for clinical laboratory medicine. Exp Mol Pathol 85:50–58. doi:10.1016/j.yexmp.2008.03.012.
    OpenUrlCrossRefPubMedWeb of Science
  167. 167.↵
    1. Gundry CN,
    2. Vandersteen JG,
    3. Reed GH,
    4. Pryor RJ,
    5. Chen J,
    6. Wittwer CT
    . 2003. Amplicon melting analysis with labeled primers: a closed-tube method for differentiating homozygotes and heterozygotes. Clin Chem 49:396–406. doi:10.1373/49.3.396.
    OpenUrlAbstract/FREE Full Text
  168. 168.↵
    1. Erali M,
    2. Palais R,
    3. Wittwer C
    . 2008. SNP genotyping by unlabeled probe melting analysis. Methods Mol Biol 429:199–206. doi:10.1007/978-1-60327-040-3_14.
    OpenUrlCrossRefPubMed
  169. 169.↵
    1. Dwight Z,
    2. Palais R,
    3. Wittwer CT
    . 2011. uMELT: prediction of high-resolution melting curves and dynamic melting profiles of PCR products in a rich Web application. Bioinformatics 27:1019–1020. doi:10.1093/bioinformatics/btr065.
    OpenUrlCrossRefPubMedWeb of Science
  170. 170.↵
    1. Drancourt M,
    2. Michel-Lepage A,
    3. Boyer S,
    4. Raoult D
    . 2016. The point-of-care laboratory in clinical microbiology. Clin Microbiol Rev 29:429–447. doi:10.1128/CMR.00090-15.
    OpenUrlAbstract/FREE Full Text
  171. 171.↵
    1. Penno EC,
    2. Crump JA,
    3. Baird SJ
    . 2015. Performance requirements to achieve cost-effectiveness of point-of-care tests for sepsis among patients with febrile illness in low-resource settings. Am J Trop Med Hyg 93:841–849. doi:10.4269/ajtmh.15-0082.
    OpenUrlAbstract/FREE Full Text
  172. 172.↵
    1. Vernon SD,
    2. Shukla SK,
    3. Conradt J,
    4. Unger ER,
    5. Reeves WC
    . 2002. Analysis of 16S rRNA gene sequences and circulating cell-free DNA from plasma of chronic fatigue syndrome and non-fatigued subjects. BMC Microbiol 2:39. doi:10.1186/1471-2180-2-39.
    OpenUrlCrossRefPubMed
  173. 173.↵
    1. Rogers GB,
    2. Marsh P,
    3. Stressmann AF,
    4. Allen CE,
    5. Daniels TVW,
    6. Carroll MP,
    7. Bruce KD
    . 2010. The exclusion of dead bacterial cells is essential for accurate molecular analysis of clinical samples. Clin Microbiol Infect 16:1656–1658. doi:10.1111/j.1469-0691.2010.03189.x.
    OpenUrlCrossRefPubMed
  174. 174.↵
    1. Jo T,
    2. Murakami H,
    3. Masuda R,
    4. Sakata MK,
    5. Yamamoto S,
    6. Minamoto T
    . 2017. Rapid degradation of longer DNA fragments enables the improved estimation of distribution and biomass using environmental DNA. Mol Ecol Resour 17:e25–e33. doi:10.1111/1755-0998.12685.
    OpenUrlCrossRef
  175. 175.↵
    1. Ziegler I,
    2. Josefson P,
    3. Olcén P,
    4. Mölling P,
    5. Strålin K
    . 2014. Quantitative data from the SeptiFast real-time PCR is associated with disease severity in patients with sepsis. BMC Infect Dis 14:155. doi:10.1186/1471-2334-14-155.
    OpenUrlCrossRef
  176. 176.↵
    1. Reers Y,
    2. Idelevich EA,
    3. Pätkau H,
    4. Sauerland MC,
    5. Tafelski S,
    6. Nachtigall I,
    7. Berdel WE,
    8. Peters G,
    9. Silling G,
    10. Becker K
    . 2016. Multiplex PCR assay underreports true bloodstream infections with coagulase-negative staphylococci in hematological patients with febrile neutropenia. Diagn Microbiol Infect Dis 85:413–415. doi:10.1016/j.diagmicrobio.2016.04.024.
    OpenUrlCrossRef
  177. 177.↵
    1. Rogina P,
    2. Skvarc M,
    3. Stubljar D,
    4. Kofol R,
    5. Kaasch A
    . 2014. Diagnostic utility of broad range bacterial 16S rRNA gene PCR with degradation of human and free bacterial DNA in bloodstream infection is more sensitive than an in-house developed PCR without degradation of human and free bacterial DNA. Mediators Inflamm 2014:108592. doi:10.1155/2014/108592.
    OpenUrlCrossRef
  178. 178.↵
    1. Mihajlovic D,
    2. Brkic S,
    3. Uvelin A,
    4. Draskovic B,
    5. Vrsajkov V
    . 2017. Use of presepsin and procalcitonin for prediction of SeptiFast results in critically ill patients. J Crit Care 40:197–201. doi:10.1016/j.jcrc.2017.04.008.
    OpenUrlCrossRef
  179. 179.↵
    1. Schreiber J,
    2. Nierhaus A,
    3. Braune SA,
    4. de Heer G,
    5. Kluge S
    . 2013. Comparison of three different commercial PCR assays for the detection of pathogens in critically ill sepsis patients. Med Klin Intensivmed Notfmed 108:311–318. doi:10.1007/s00063-013-0227-1.
    OpenUrlCrossRefPubMed
  180. 180.↵
    1. Wilson IG
    . 1997. Inhibition and facilitation of nucleic acid amplification. Appl Environ Microbiol 63:3741–3751.
    OpenUrlFREE Full Text
  181. 181.↵
    1. Rantakokko-Jalava K,
    2. Jalava J
    . 2002. Optimal DNA isolation method for detection of bacteria in clinical specimens by broad-range PCR. J Clin Microbiol 40:4211–4217. doi:10.1128/JCM.40.11.4211-4217.2002.
    OpenUrlAbstract/FREE Full Text
  182. 182.↵
    1. Morata P,
    2. Queipo-Ortuño MI,
    3. de Dios Colmenero J
    . 1998. Strategy for optimizing DNA amplification in a peripheral blood PCR assay used for diagnosis of human brucellosis. J Clin Microbiol 36:2443–2446.
    OpenUrlAbstract/FREE Full Text
  183. 183.↵
    1. Tanner NA,
    2. Zhang Y,
    3. Evans TC, Jr
    . 2012. Simultaneous multiple target detection in real-time loop-mediated isothermal amplification. Biotechniques 53:81–89.
    OpenUrlCrossRefPubMed
  184. 184.↵
    1. Connelly JT,
    2. Rolland JP,
    3. Whitesides GM
    . 2015. “Paper machine” for molecular diagnostics. Anal Chem 87:7595–7601. doi:10.1021/acs.analchem.5b00411.
    OpenUrlCrossRef
  185. 185.↵
    1. Njiru ZK
    . 2012. Loop-mediated isothermal amplification technology: towards point of care diagnostics. PLoS Negl Trop Dis 6:e1572. doi:10.1371/journal.pntd.0001572.
    OpenUrlCrossRefPubMed
  186. 186.↵
    1. Mchugh L,
    2. Seldon TA,
    3. Brandon RA,
    4. Kirk JT,
    5. Rapisarda A,
    6. Sutherland AJ,
    7. Presneill JJ,
    8. Venter DJ,
    9. Lipman J,
    10. Thomas MR,
    11. Klein Klouwenberg PM,
    12. van Vught L,
    13. Scicluna B,
    14. Bonten M,
    15. Cremer OL,
    16. Schultz MJ,
    17. van der Poll T,
    18. Yager TD,
    19. Brandon RB
    . 2015. A molecular host response assay to discriminate between sepsis and infection-negative systemic inflammation in critically ill patients: discovery and validation in independent cohorts. PLoS Med 12:e1001916. doi:10.1371/journal.pmed.1001916.
    OpenUrlCrossRefPubMed
  187. 187.↵
    1. Zimmerman JJ,
    2. Sullivan E,
    3. Yager TD,
    4. Cheng C,
    5. Permut L,
    6. Cermelli S,
    7. McHugh L,
    8. Sampson D,
    9. Seldon T,
    10. Brandon RB,
    11. Brandon RA
    . 2017. Diagnostic accuracy of a host gene expression signature that discriminates clinical severe sepsis syndrome and infection-negative systemic inflammation among critically ill children. Crit Care Med 45:e418–e425. doi:10.1097/CCM.0000000000002100.
    OpenUrlCrossRef
  188. 188.↵
    1. Zimmerman J,
    2. Sullivan E,
    3. Sampson D,
    4. McHugh L,
    5. Yager T,
    6. Seldon T
    . 2015. Sensitive and specific diagnosis of sepsis in critically ill children utilizing host gene expression. Crit Care Med 43:258. doi:10.1097/01.ccm.0000474855.13970.46.
    OpenUrlCrossRef
  189. 189.↵
    1. Miller R,
    2. Lopansri B,
    3. McHugh L,
    4. Rapisarda A,
    5. Seldon T,
    6. Burke J
    . 2015. Validation of a novel host response assay to distinguish SIRS and sepsis in critically ill patients. Crit Care Med 43:252. doi:10.1097/01.ccm.0000474833.48035.b5.
    OpenUrlCrossRef
  190. 190.↵
    1. Kang D-K,
    2. Ali MM,
    3. Zhang K,
    4. Huang SS,
    5. Peterson E,
    6. Digman MA,
    7. Gratton E,
    8. Zhao W
    . 2014. Rapid detection of single bacteria in unprocessed blood using integrated comprehensive droplet digital detection. Nat Commun 5:5427. doi:10.1038/ncomms6427.
    OpenUrlCrossRef
  191. 191.↵
    1. Skinner JP,
    2. Swift KM,
    3. Ruan Q,
    4. Perfetto S,
    5. Gratton E,
    6. Tetin SY
    . 2013. Simplified confocal microscope for counting particles at low concentrations. Rev Sci Instrum 84:74301. doi:10.1063/1.4812782.
    OpenUrlCrossRef
  192. 192.↵
    1. Altamore I,
    2. Lanzano L,
    3. Gratton E
    . 2013. Dual channel detection of ultra low concentration of bacteria in real time by scanning FCS. Meas Sci Technol 24:65702. doi:10.1088/0957-0233/24/6/065702.
    OpenUrlCrossRef
  193. 193.
    Reference deleted.
  194. 194.↵
    1. Toll DB,
    2. Janssen KJM,
    3. Vergouwe Y,
    4. Moons KGM
    . 2008. Validation, updating and impact of clinical prediction rules: a review. J Clin Epidemiol 61:1085–1094. doi:10.1016/j.jclinepi.2008.04.008.
    OpenUrlCrossRefPubMedWeb of Science
  195. 195.↵
    1. Semler MW,
    2. Weavind L,
    3. Hooper MH,
    4. Rice TW,
    5. Gowda SS,
    6. Nadas A,
    7. Song Y,
    8. Martin JB,
    9. Bernard GR,
    10. Wheeler AP
    . 2015. An electronic tool for the evaluation and treatment of sepsis in the ICU. Crit Care Med 43:1595–1602. doi:10.1097/CCM.0000000000001020.
    OpenUrlCrossRefPubMed
  196. 196.↵
    1. Hooper MH,
    2. Weavind L,
    3. Wheeler AP,
    4. Martin JB,
    5. Gowda SS,
    6. Semler MW,
    7. Hayes RM,
    8. Albert DW,
    9. Deane NB,
    10. Nian H,
    11. Mathe JL,
    12. Nadas A,
    13. Sztipanovits J,
    14. Miller A,
    15. Bernard GR,
    16. Rice TW
    . 2012. Randomized trial of automated, electronic monitoring to facilitate early detection of sepsis in the intensive care unit. Crit Care Med 40:2096–2101. doi:10.1097/CCM.0b013e318250a887.
    OpenUrlCrossRefPubMedWeb of Science
  197. 197.↵
    1. Umscheid CA,
    2. Betesh J,
    3. VanZandbergen C,
    4. Hanish A,
    5. Tait G,
    6. Mikkelsen ME,
    7. French B,
    8. Fuchs BD
    . 2015. Development, implementation, and impact of an automated early warning and response system for sepsis. J Hosp Med 10:26–31. doi:10.1002/jhm.2259.
    OpenUrlCrossRefPubMed
  198. 198.↵
    1. Pierrakos C,
    2. Vincent J-L
    . 2010. Sepsis biomarkers: a review. Crit Care 14:R15. doi:10.1186/cc8872.
    OpenUrlCrossRefPubMed
  199. 199.↵
    1. Taneja I,
    2. Reddy B,
    3. Damhorst G,
    4. Zhao SD,
    5. Hassan U,
    6. Price Z,
    7. Jensen T,
    8. Ghonge T,
    9. Patel M,
    10. Wachspress S,
    11. Winter J,
    12. Rappleye M,
    13. Smith G,
    14. Healey R,
    15. Ajmal M,
    16. Khan M,
    17. Patel J,
    18. Rawal H,
    19. Sarwar R,
    20. Soni S,
    21. Anwaruddin S,
    22. Davis B,
    23. Kumar J,
    24. White K,
    25. Bashir R,
    26. Zhu R
    . 2017. Combining biomarkers with EMR data to identify patients in different phases of sepsis. Sci Rep 7:10800. doi:10.1038/s41598-017-09766-1.
    OpenUrlCrossRef
  200. 200.↵
    1. Liu Y,
    2. Choi KS
    . 2017. Using machine learning to diagnose bacterial sepsis in the critically ill patients, p 223–233. In International Conference on Smart Health 2017. Springer, Cham, Switzerland. doi:10.1007/978-3-319-67964-8_22.
    OpenUrlCrossRef
  201. 201.↵
    1. Wong HR,
    2. Weiss SL,
    3. Giuliano JS,
    4. Wainwright MS,
    5. Cvijanovich NZ,
    6. Thomas NJ,
    7. Allen GL,
    8. Anas N,
    9. Bigham MT,
    10. Hall M,
    11. Freishtat RJ,
    12. Sen A,
    13. Meyer K,
    14. Checchia PA,
    15. Shanley TP,
    16. Nowak J,
    17. Quasney M,
    18. Chopra A,
    19. Fitzgerald JC,
    20. Gedeit R,
    21. Banschbach S,
    22. Beckman E,
    23. Harmon K,
    24. Lahni P,
    25. Lindsell CJ
    . 2014. The temporal version of the pediatric sepsis biomarker risk model. PLoS One 9:e92121. doi:10.1371/journal.pone.0092121.
    OpenUrlCrossRefPubMed
  202. 202.↵
    1. Henry KE,
    2. Hager DN,
    3. Pronovost PJ,
    4. Saria S
    . 2015. A targeted real-time early warning score (TREWScore) for septic shock. Sci Transl Med 7:299ra122. doi:10.1126/scitranslmed.aab3719.
    OpenUrlAbstract/FREE Full Text
  203. 203.↵
    1. Subbe CP,
    2. Slater A,
    3. Menon D,
    4. Gemmell L
    . 2006. Validation of physiological scoring systems in the accident and emergency department. Emerg Med J 23:841–845. doi:10.1136/emj.2006.035816.
    OpenUrlAbstract/FREE Full Text
  204. 204.↵
    1. Mani S,
    2. Ozdas A,
    3. Aliferis C,
    4. Varol HA,
    5. Chen Q,
    6. Carnevale R,
    7. Chen Y,
    8. Romano-Keeler J,
    9. Nian H,
    10. Weitkamp J-H
    . 2014. Medical decision support using machine learning for early detection of late-onset neonatal sepsis. J Am Med Inform Assoc 21:326–336. doi:10.1136/amiajnl-2013-001854.
    OpenUrlCrossRefPubMed
  205. 205.↵
    1. Wang K,
    2. Bhandari V,
    3. Chepustanova S,
    4. Huber G,
    5. O'Hara S,
    6. O'Hern CS,
    7. Shattuck MD,
    8. Kirby M
    . 2013. Which biomarkers reveal neonatal sepsis? PLoS One 8:e82700. doi:10.1371/journal.pone.0082700.
    OpenUrlCrossRefPubMed
  206. 206.↵
    1. Mayhew MB,
    2. Petersen BK,
    3. Sales AP,
    4. Greene JD,
    5. Liu VX,
    6. Wasson TS
    . 2018. Flexible, cluster-based analysis of the electronic medical record of sepsis with composite mixture models. J Biomed Inform 78:33–42. doi:10.1016/j.jbi.2017.11.015.
    OpenUrlCrossRef
  207. 207.↵
    1. Moorman JR,
    2. Carlo WA,
    3. Kattwinkel J,
    4. Schelonka RL,
    5. Porcelli PJ,
    6. Navarrete CT,
    7. Bancalari E,
    8. Aschner JL,
    9. Walker MW,
    10. Perez JA,
    11. Palmer C,
    12. Stukenborg GJ,
    13. Lake DE,
    14. O'Shea TM
    . 2011. Mortality reduction by heart rate characteristic monitoring in very low birth weight neonates: a randomized trial. J Pediatr 159:900.e1–906.e1. doi:10.1016/j.jpeds.2011.06.044.
    OpenUrlCrossRef
  208. 208.↵
    1. Sullivan BA,
    2. Grice SM,
    3. Lake DE,
    4. Moorman JR,
    5. Fairchild KD
    . 2014. Infection and other clinical correlates of abnormal heart rate characteristics in preterm infants. J Pediatr 164:775–780. doi:10.1016/j.jpeds.2013.11.038.
    OpenUrlCrossRefPubMed
  209. 209.↵
    1. Coggins SA,
    2. Weitkamp J-H,
    3. Grunwald L,
    4. Stark AR,
    5. Reese J,
    6. Walsh W,
    7. Wynn JL
    . 2016. Heart rate characteristic index monitoring for bloodstream infection in an NICU: a 3-year experience. Arch Dis Child Fetal Neonatal Ed 101:F329–F332. doi:10.1136/archdischild-2015-309210.
    OpenUrlAbstract/FREE Full Text
  210. 210.↵
    1. Desautels T,
    2. Calvert J,
    3. Hoffman J,
    4. Jay M,
    5. Kerem Y,
    6. Shieh L,
    7. Shimabukuro D,
    8. Chettipally U,
    9. Feldman MD,
    10. Barton C,
    11. Wales DJ,
    12. Das R
    . 2016. Prediction of sepsis in the intensive care unit with minimal electronic health record data: a machine learning approach. JMIR Med Inform 4:e28. doi:10.2196/medinform.5909.
    OpenUrlCrossRef
  211. 211.↵
    1. Calvert J,
    2. Desautels T,
    3. Chettipally U,
    4. Barton C,
    5. Hoffman J,
    6. Jay M,
    7. Mao Q,
    8. Mohamadlou H,
    9. Das R
    . 2016. High-performance detection and early prediction of septic shock for alcohol-use disorder patients. Ann Med Surg 8:50–55. doi:10.1016/j.amsu.2016.04.023.
    OpenUrlCrossRef
  212. 212.↵
    1. Calvert JS,
    2. Price DA,
    3. Chettipally UK,
    4. Barton CW,
    5. Feldman MD,
    6. Hoffman JL,
    7. Jay M,
    8. Das R
    . 2016. A computational approach to early sepsis detection. Comput Biol Med 74:69–73. doi:10.1016/j.compbiomed.2016.05.003.
    OpenUrlCrossRef
  213. 213.↵
    1. Desautels T,
    2. Hoffman J,
    3. Barton C,
    4. Mao Q,
    5. Jay M,
    6. Calvert J,
    7. Das R
    . 2017. Pediatric severe sepsis prediction using machine learning. bioRxiv doi:10.1101/223289.
    OpenUrlAbstract/FREE Full Text
  214. 214.↵
    1. Calvert JS,
    2. Price DA,
    3. Barton CW,
    4. Chettipally UK,
    5. Das R
    . 2017. Discharge recommendation based on a novel technique of homeostatic analysis. J Am Med Inform Assoc 24:24–29. doi:10.1093/jamia/ocw014.
    OpenUrlCrossRef
  215. 215.↵
    1. McCoy A,
    2. Das R
    . 2017. Reducing patient mortality, length of stay and readmissions through machine learning-based sepsis prediction in the emergency department, intensive care unit and hospital floor units. BMJ Open Qual 6:e000158. doi:10.1136/bmjoq-2017-000158.
    OpenUrlAbstract/FREE Full Text
  216. 216.↵
    1. Calvert J,
    2. Hoffman J,
    3. Barton C,
    4. Shimabukuro D,
    5. Ries M,
    6. Chettipally U,
    7. Kerem Y,
    8. Jay M,
    9. Mataraso S,
    10. Das R
    . 2017. Cost and mortality impact of an algorithm-driven sepsis prediction system. J Med Econ 20:646–651. doi:10.1080/13696998.2017.1307203.
    OpenUrlCrossRef

Author Bios


Embedded Image

Mridu Sinha, M.S., is a Bioengineering doctoral candidate at the University of California, San Diego. Ms. Sinha received her bachelor of science degree in Electrical Engineering from Manipal University and her master of science degree in Electrical Engineering from University of Wisconsin—Madison. Her graduate work has been focused on understanding the challenges and developing solutions to provide timely care for newborns with asphyxia and sepsis. Through her graduate work, Ms. Sinha has developed an understanding of the existing and emerging technologies and gaps that remain to be addressed in sepsis diagnostics.


Embedded Image

Julietta Jupe, Ph.D., is currently conducting postdoctoral research at the Donald Danforth Plant Science Center. At the time when the manuscript was being prepared, Dr. Jupe was a postdoctoral fellow at the University of California, San Diego, under the guidance of Dr. Stephanie Fraley. Dr. Jupe received her engineer's degree in Horticulture from Fachhochschule Weibhenstephan Freising and her Ph.D. in Molecular Microbiology from The University of Dundee.


Embedded Image

Hannah Mack, M.S., is a Technology Development Researcher at the University of California, San Diego. She received her bachelor of science degree in Mechanical Engineering from the University of Dayton and her master of science degree in Bioengineering from the University of California, San Diego. Ms. Mack codeveloped the current U-dHRM platform with Ms. Sinha as her thesis project and has extensive expertise in instrumentation and assay development specific to this project. Prior to graduate school, Ms. Mack also worked at a small lower-limb prosthetics company, where she manufactured custom prosthetic parts and gained invaluable patient experience.


Embedded Image

Todd P. Coleman, Ph.D., is a Professor in the Department of Bioengineering at the University of California, San Diego. Dr. Coleman received his bachelor of science degrees in electrical engineering and computer engineering from the University of Michigan and his master of science and Ph.D. degrees in electrical engineering at MIT under the guidance of Muriel Médard. His postdoctoral fellowship in neuroscience at Mass General Hospital and MIT was conducted under the guidance of Emery Brown. Dr. Coleman was also an Assistant Professor of Electrical and Computer Engineering and Neuroscience at the University of Illinois, Urbana-Champaign, from 2006 until 2011.


Embedded Image

Shelley M. Lawrence, M.D., M.S., is a neonatologist at Rady Children's Hospital of San Diego and the University of California, San Diego. She is an assistant clinical professor of pediatrics and the Neonatal Perinatal Fellowship Director. After completing her neonatal fellowship in 2004 at the University of Oklahoma Health Sciences Center, she accepted a position with the Pediatrix Medical Group of Colorado, where she worked in private-practice neonatology for 5 1/2 years. She returned to the University of Oklahoma in 2010 to practice academic medicine. At Children's Hospital at the OU Medical Center, she was codirector of the Neonatal Congenital Diaphragmatic Hernia Program and extracorporeal membrane oxygenation (ECMO) educator for neonatal fellows. Dr. Lawrence received a master of science degree in clinical and translational medicine in 2013 with a focus on neonatal neutrophil physiology. Her research interests include neutrophil function in neonatal sepsis, the development of technology for the rapid identification of sepsis, and the use of flow cytometry to evaluate dynamic changes in neutrophil composition in health and disease.


Embedded Image

Stephanie I. Fraley, Ph.D., earned her bachelor of science degree in Chemical Engineering from the University of Tennessee, Chattanooga, and her Ph.D. in Chemical and Biomolecular Engineering from The Johns Hopkins University. She then joined the Emergency Medicine and Biomedical Engineering departments at The Johns Hopkins University as a postdoctoral fellow. Dr. Fraley is now an Assistant Professor in the Department of Bioengineering at the University of California, San Diego. One of her primary research focuses is the development of novel approaches to diagnose and monitor diseases towards quantitative personalized medicine. For her 10 years of work at the interface of medicine and engineering, she has been recognized with a National Burroughs Wellcome Fund Career Award at the Scientific Interface and named a SAGE Bionetworks Scholar, Kavli Frontiers of Science Fellow, Biomedical Engineering Society Rising Star in Cellular and Molecular Bioengineering, and NSF CAREER awardee.

PreviousNext
Back to top
Download PDF
Citation Tools
Emerging Technologies for Molecular Diagnosis of Sepsis
Mridu Sinha, Julietta Jupe, Hannah Mack, Todd P. Coleman, Shelley M. Lawrence, Stephanie I. Fraley
Clinical Microbiology Reviews Feb 2018, 31 (2) e00089-17; DOI: 10.1128/CMR.00089-17

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Print

Alerts
Sign In to Email Alerts with your Email Address
Email

Thank you for sharing this Clinical Microbiology Reviews article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Emerging Technologies for Molecular Diagnosis of Sepsis
(Your Name) has forwarded a page to you from Clinical Microbiology Reviews
(Your Name) thought you would be interested in this article in Clinical Microbiology Reviews.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Emerging Technologies for Molecular Diagnosis of Sepsis
Mridu Sinha, Julietta Jupe, Hannah Mack, Todd P. Coleman, Shelley M. Lawrence, Stephanie I. Fraley
Clinical Microbiology Reviews Feb 2018, 31 (2) e00089-17; DOI: 10.1128/CMR.00089-17
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Top
  • Article
    • SUMMARY
    • INTRODUCTION
    • THE IDEAL SEPSIS DIAGNOSTIC TEST
    • LIMITATIONS OF STANDARD DIAGNOSTIC BLOOD CULTURE METHODOLOGIES (“GOLD STANDARD”)
    • TOWARDS DETECTION DIRECTLY FROM WHOLE BLOOD: CURRENT AND EMERGING TECHNOLOGIES FOR RAPID DIAGNOSIS OF MICROBIAL INFECTIONS WITHOUT CULTURE
    • CONCLUSION
    • ACKNOWLEDGMENTS
    • REFERENCES
    • Author Bios
  • Figures & Data
  • Info & Metrics
  • PDF

KEYWORDS

biomedical engineering
DNA sequencing
diagnostic
infectious disease
microbiology techniques

Related Articles

Cited By...

About

  • About CMR
  • Editor in Chief
  • Editorial Board
  • Policies
  • For the Media
  • For Librarians
  • For Advertisers
  • Alerts
  • RSS
  • FAQ
  • Permissions
  • Journal Announcements

Authors

  • ASM Author Center
  • Submit a Manuscript
  • Ethics
  • Contact Us

Follow #ClinMicroRev

@ASMicrobiology

       

ASM Journals

ASM journals are the most prominent publications in the field, delivering up-to-date and authoritative coverage of both basic and clinical microbiology.

About ASM | Contact Us | Press Room

 

ASM is a member of

Scientific Society Publisher Alliance

 

American Society for Microbiology
1752 N St. NW
Washington, DC 20036
Phone: (202) 737-3600

Copyright © 2021 American Society for Microbiology | Privacy Policy | Website feedback

Print ISSN: 0893-8512; Online ISSN: 1098-6618