Metagenomic sequencing for infectious disease diagnostics is an important tool that holds promise for use in the clinical laboratory. Challenges for implementation so far include high cost, the length of time to results, and the need for technical and bioinformatics expertise. However, the recent technological innovation of nanopore sequencing from Oxford Nanopore Technologies (ONT) has the potential to address these challenges.
KEYWORDS: nanopore sequencing, third-generation sequencing, infectious disease diagnostics
ABSTRACT
Metagenomic sequencing for infectious disease diagnostics is an important tool that holds promise for use in the clinical laboratory. Challenges for implementation so far include high cost, the length of time to results, and the need for technical and bioinformatics expertise. However, the recent technological innovation of nanopore sequencing from Oxford Nanopore Technologies (ONT) has the potential to address these challenges. ONT sequencing is an attractive platform for clinical laboratories to adopt due to its low cost, rapid turnaround time, and user-friendly bioinformatics pipelines. However, this method still faces the problem of base-calling accuracy compared to other platforms. This review highlights the general challenges of pathogen detection in clinical specimens by metagenomic sequencing, the advantages and disadvantages of the ONT platform, and how research to date supports the potential future use of nanopore sequencing in infectious disease diagnostics.
INTRODUCTION
Infectious disease diagnostics currently involve an array of laboratory methods, including culture, serologic assays, nucleic acid amplification tests, antigen detection, and direct visualization. The complexity of diagnostic options requires nuanced understanding on the part of the clinician and careful assessment of a patient’s clinical presentation and history to ensure appropriate test ordering. For culture-based methods, a growth amplification step is necessary that can take anywhere from a day to several weeks, depending on the type of pathogen causing infection. The likelihood of isolating a pathogen is compromised when fastidious organisms are present or when a patient is receiving antimicrobial therapy. Developing increasingly rapid, accurate methods to identify pathogens and characterize antimicrobial resistance (AMR) is a constant quest in the field of infectious disease diagnostics in order to improve patient outcomes. In the face of the complexity of current methods, metagenomic next-generation sequencing (NGS) offers the possibility of universal pathogen detection, enabling the identification of bacteria, fungi, viruses, and parasites with a single method directly from patient specimens (1, 2).
The advent of NGS in the early 2000s revolutionized the field of genomic research. NGS encompasses several different approaches to nucleic acid sequencing; however, they all utilize the same basic approach in which either DNA or RNA molecules are sequenced in a massively parallel manner (3). The initial preparation of samples for sequencing is often a technically laborious process wherein DNA or RNA has to be isolated, checked for quality metrics, and then put through a library preparation protocol that can take hours to days to complete. Sequencing itself can then take anywhere from 1 to 6 days, depending on the platform used, the length of the reads, and the amount of data generated (Table 1). Illumina and Thermo Fisher offer short-read (100 to 400 bp) sequencing platforms, whereas Pacific Biosciences and Oxford Nanopore Technologies (ONT) offer long-read (>500 bp) sequencing (3, 4). Each platform offers its own advantages and disadvantages in terms of accuracy, efficiency, and cost. Regardless of the technology used, bioinformatics knowledge is required for data processing and analysis. Furthermore, turnaround time from nucleic acid extraction through final result reporting typically takes a minimum of 5 to 10 days.
TABLE 1.
Sequencing platform | Chemistry | Avg read length (bp) | Advantage(s) | Disadvantage(s)a |
---|---|---|---|---|
Illumina | Sequencing by synthesis; fluorescently labeled deoxynucleoside triphosphates | ≤300 | High accuracy | Short reads, high capital cost, long TAT |
Thermo Fisher Ion Torrent | Sequencing by synthesis; detection of hydrogen ions | ≤400 | High accuracy | Short reads, high capital cost, long TAT |
Pacific Biosciences | Sequencing by synthesis: SMRTbell replication | ≥500 | Long reads | High capital cost, variable accuracy, long TAT |
Oxford Nanopore | Measures the changes in current as biological molecules pass through the nanopore | ≥500 | Long reads, low capital cost, short TAT | Low accuracy |
TAT, turnaround time.
NGS in the clinical setting has become widespread in personalized medicine due to its ability to characterize variants throughout the human genome. The Illumina and Thermo Fisher platforms are in widespread use to identify somatic mutations in cancer and to uncover the genetic basis of various inherited diseases (5, 6). Application of NGS to infectious disease diagnostics has been slower to evolve for a number of reasons. In this review, we focus on challenges for implementation of metagenomic sequencing for pathogen detection, why nanopore sequencing could overcome many of those hurdles, and how research performed to date supports the future use of ONT sequencing in the clinical laboratory.
NGS FOR INFECTIOUS DISEASE DIAGNOSTICS
The use of NGS for infectious disease diagnostics is being increasingly explored and adopted for select applications in the clinical laboratory (4, 7–12). Several approaches to pathogen detection with NGS can be undertaken (Table 2). Metagenomic whole-genome sequencing (mWGS), which analyzes all of the DNA or RNA in a given sample, offers an unbiased, hypothesis-free approach to detecting bacteria, viruses, fungi, and parasites. Aside from high cost, an additional consideration for performing mWGS is the abundance of human DNA in many clinical sample types such as blood and respiratory secretions (13, 14). Human contamination results in most sequence data belonging to the host instead of pathogen, making it more difficult to detect and characterize infectious agents. Furthermore, analysis of any samples containing human reads should be done on a secure, HIPAA (Health Insurance Portability and Accountability Act of 1996) compliant platform to ensure protection of genomic information. An alternative approach for bacterial pathogens is targeted, 16S rRNA gene sequencing (15, 16). The 16S rRNA gene has been the most commonly used region for bacterial detection and identification because it exists in almost all bacteria, because it has conserved regions that enable universal primer design, and because smaller portions of the gene, termed variable regions, can be sequenced, as opposed to the entire gene (1,500 bp) (17). Polymorphisms within the variable regions are what distinguish different species of bacteria with <2% variability differentiating closely related species (18). Similarly, internal transcribed spacer (ITS) sequencing can be utilized to target fungi for sequencing (19). Advantages of targeted methods over mWGS include increased sensitivity, decreased cost, and decreased complexity of computationally intensive bioinformatics. However, 16S rRNA and ITS sequencing primers will not pick up every bacterial or fungal species due to variation in nucleotide composition at primer binding sites (19, 20). In addition, 16S rRNA gene and ITS sequencing can provide only taxonomic identification while mWGS can also provide genomic data on virulence, AMR, metabolism, and strain typing that may be of clinical or epidemiologic utility.
TABLE 2.
Sequencing approach | Target | Advantage(s) | Disadvantage(s) |
---|---|---|---|
16S rRNA gene | Bacteria | Lower cost, targets and amplifies most bacteria present, low-complexity bioinformatics | Specific to bacteria, can still miss some species due to primer mismatches, bacterial ID only |
18S rRNA gene/ITS gene | Fungi and some parasites | Lower cost, targets and amplifies most fungi present and some parasites, low-complexity bioinformatics | Specific to fungi and a subset of parasites, can still miss some species due to primer mismatches |
Whole genome | Cultured organism | Provides in-depth coverage of a single organism with associated virulence and antimicrobial genes | Requires a cultured organism, high-complexity bioinformatics |
Metagenomic whole genome | Unbiased | Sequence any organism that is present, can analyze entire genomes, ID of virulence and AMR genes | High cost, high-complexity bioinformatics, host contamination |
ID, identification.
To date, regardless of the sequencing approach, logistical hurdles for implementation include the high cost for capital investment and consumables, availability of trained/experienced laboratory personnel and lack of prospective clinical outcomes data to justify increased laboratory costs. In addition, there are numerous possible approaches to wet lab work flow all of which must be optimized, including storage and processing, nucleic acid isolation protocols, controlling for environmental contamination, sequencing library preparation methods, and choice of sequencing platform. The wet lab protocols to be used are highly dependent on the sample type, the type of organism(s) targeted, the amount of nucleic acid isolated, and the sequencing approach. The overall design of each step in the process can influence final results and even the ability to detect certain pathogens (21).
In addition to the variability of wet lab protocols, the bioinformatics for data handling and interpretation can be resource intense. Bioinformatic analysis requires highly trained staff, valid analysis tools, including the reference database, the computational infrastructure, and the creation of standardized procedures. Each of the four sequencing platforms (Illumina, Thermo Fisher, Pacific Biosciences, and ONT) require their own initial data processing steps and quality control metrics. For bioinformatics, the tools that have been developed in the research community for short-read data are not feasible for long-read data. Data interpretation adds an additional level of complexity. In clinical specimens it can at times be difficult to distinguish normal microbiota or colonizers from pathogens since certain organisms can be either (1). Thresholds of detection for clinical relevance for certain organisms in certain specimen types need to be established before routine use of NGS for common specimen types is possible. The true complexities of how different bioinformatics pipelines influence data analysis is beyond the scope of this review; however, there is no one “correct” way to handle sequence data.
Due to the multitude of options available for everything from sample collection through final interpretation of results, efforts are under way to standardize key parts of the process for NGS in infectious disease diagnostics. The recent release of the FDA-ARGOS reference database signified major headway in the direction of standardization (22). However, a plethora of challenges remain for how to establish protocols from sample collection all the way through final data analysis (7). It is likely that standardized protocols will need to be developed for each sample type, sequencing approach, and sequencing platform. The inclusion of negative and/or no template controls should be mandatory to aid in identifying any possible false-positive results. One of the few commonalities across platforms and sample types will be the database used and specific thresholds (such as read depth and genome coverage) used during data analysis, parameters which are already being explored (10, 11). Also important for validation across different laboratories will be proficiency material, such as the use of commercial standards from companies such as the American Type Culture Collection that contain mixed microbial communities.
Another barrier to implementation of NGS in infectious disease diagnostics has been the time it takes to complete all the steps for NGS. In general, it can take a minimum of 1 week to prepare samples, sequence libraries, and analyze data once standardized protocols are put into place. Altogether, this turnaround time relative to conventional methods has limited the clinical relevance of NGS results for patient care decision-making. Because of this, many NGS applications in the clinical microbiology laboratory target situations where a rapid answer is not necessary. This includes outbreak surveillance, infection prevention, and WGS of antibiotic-resistant isolates (23–27). Rapidly evolving NGS technology, however, may soon address many of the challenges discussed thus far.
NANOPORE SEQUENCING
While NGS platforms from Illumina, Thermo Fisher, and Pacific Biosciences have revolutionized biomedical research, a novel approach to NGS using nanopore technology has the potential for use in the clinical lab in the near future. Oxford Nanopore Technologies (ONT) first introduced the MinION platform to the research market in 2014. MinION is different from other platforms because it utilizes nanopores for sequencing (28). It does not take a sequencing-by-synthesis approach; instead, an ionic current is passed across the flow cell during sequencing, and the different nucleotide bases are distinguished by the changes in current as they pass through the nanopores (28). Sequencing with the MinION platform requires minimal capital cost compared to the Illumina, Thermo Fisher, and Pacific Biosciences platforms and can be utilized both in the laboratory and out in the field. Able to fit in the palm of your hand and connected to either ONT’s “MinIT” computer module or to any computer with a USB connection, MinION permits direct, electronic analysis of single molecules in real time. It can be used to analyze DNA and RNA for a range of applications, including personalized medicine, agriculture, and scientific research.
Although nanopore sequencing is able to produce reads of up to 2 Mb in length (29), the biggest drawbacks to date have been a lower throughput of sequence data and a high error rate (approximately 10%) with their 9.4 and earlier-version flow cells that use 1D chemistry (30). The 9.5 flow cells utilize 1D2 chemistry and have been reported to achieve 99% accuracy in base calling of the 16S rRNA gene of a group of commonly identified sepsis-causing organisms (31). Furthermore, the new version 10 flow cells utilize a new type of nanopore, and ONT claims this flow cell can deliver up to 99.99% base-calling accuracy. Regardless of these recent improvements, the lower accuracy in base calling with earlier versions of the MinION flow cells has not limited the use of nanopore sequencing in the infectious disease research setting. Validation of the accuracy of nanopore sequencing long reads has been achieved by the inclusion of parallel sequencing with Illumina. In addition, long reads generated by ONT have been used to fill in the gaps in unfinished genomes sequenced on short-read platforms (32–37).
The advantages of nanopore long-read sequencing are numerous. No other platform allows for real-time analysis while sequencing is ongoing, which has been one of the major drawbacks for infectious disease diagnostics with other types of NGS technology. A MinION flow cell can accommodate numerous concurrent patient samples and generates between 4 and 8 GB of data. It can then be reused up to ten times to maximize use of a single flow cell, which in turn can lower costs. This is advantageous for instances when only a few samples are available for sequencing and/or low read depth is required. Another advantage of nanopore sequencing is the time required for sequencing library preparation. If enough DNA is available (400 ng in 7.5 μl) the Oxford Nanopore rapid barcoding kit allows for a 10-min library preparation before loading the flow cell. Performing a sequencing run and obtaining primary acquisition of data are done with the graphical user interface program MinKNOW. MinKNOW is used for selecting run parameters, tracking platform chemistry, and producing FAST5 files (raw signal data files). Users have the option of using MinKNOW for also producing FASTQ files, a method which then uses a data processing toolkit called Guppy to convert FAST5 files to FASTQ.
Alternatively, if users have bioinformatics expertise, there are a number of tools available for data analysis that aid in improving base-calling accuracy (38). The Guppy toolkit can be downloaded separately and used to process FAST5 files through command line interface on their computer. Utilizing Guppy in this manner offers the user several algorithms for base calling, which can improve raw read accuracy by upwards of 3%. The Guppy toolkit also includes downstream analysis components to allow for demultiplexing, adapter trimming, and alignment. There are also a number of higher complexity tools available for users looking to do more than simple taxonomic classification. Those developed by ONT can be found on their github page (https://github.com/nanoporetech). Available tools, among others, include medaka, tombo, pomoxis, and nanopolish, which are used for sequence correction, identifying modified nucleotides from raw sequencing data, genome assembly, and error correction in genomic assemblies, respectively (39–42).
The stated goal of ONT has been to enable the analysis of any living thing, by any person, in any environment. To support that goal, ONT created Metrichor, a branch of the company which has developed graphical user interface tools as part of their EPI2ME platform for users with limited bioinformatics knowledge (https://epi2me.nanoporetech.com). The apps under EPI2ME allow users to perform a number of different analyses, including demultiplexing, adapter trimming, and alignment to reference genomes. Additional options for microbiological specimens include taxonomic classification (using the “What’s in my pot?” app), AMR gene ID, and direct alignment to a reference genome. Overall, these apps allow a user of ONT sequencing to process their data without needing to run software through the command line interface on their computer, an option not readily available from other platforms.
In the clinical laboratory, the most advantageous and readily applied use of nanopore sequencing will likely be in infectious disease diagnostics. As already discussed, it is an appealing option due to rapid library preparation and sequencing and user-friendly bioinformatics. Further addressing concerns over the cost for NGS, ONT is working to reduce the cost of a sequencing run with the introduction of the Flongle in March of 2019. The Flongle is an adapter (flow cell dongle) for MinION that performs sequencing on smaller, single-use flow cells. At a cost of approximately $100, it is significantly less expensive than the MinION flow cells that currently cost $900. Sequencing with the Flongle allows the user to generate approximately 1 GB of data in approximately 24 h and is therefore a cost-efficient and rapid option for smaller sequencing experiments. This is ideal for infectious disease diagnostics where optimizing antimicrobial therapy depends on rapid turnaround time for pathogen detection, identification and AMR characterization.
If a user in a clinical laboratory wants to perform taxonomic classification to a more expanded and secure database than the “What’s in my pot?” app in EPI2ME, an alternative fast and easy option to explore is the One Codex platform (One Codex, San Francisco, CA). One Codex is an online platform that offers taxonomic analysis, AMR prediction, and direct alignments for any microbiology-based samples (43). The One Codex general database includes >61,000 bacterial, >48,000 viral, >1,800 fungal, >1,900 archaeal, and >200 protozoan genomes. It offers a targeted locus database for 16S, 5S, 23S, gyrB, rpoB, 18S, 28S, and ITS gene analysis. The AMR panel includes >200 genes, and the analysis includes percent identity, percent coverage, and read depth to each gene. One Codex also offers a HIPAA-compliant account, making it a better option than analysis with EPI2ME apps, which do not offer the level of security necessary for uploading patient samples. One Codex is one possible option for fast, secure analysis for laboratories who do not want to invest the time or resources for in-house microbial classification pipelines. Other commercially available platforms for bioinformatic analysis, such as Diversigen (44), CosmosID (45), and Real Time Genomics (46), are additional options for clinical laboratories unable to build and maintain taxonomic databases.
NANOPORE SEQUENCING IN INFECTIOUS DISEASE RESEARCH
Despite several hurdles that ONT still faces, a number of research publications have illustrated just how versatile and applicable nanopore sequencing is. Not only could the MinION be taken directly to the bedside of a patient, nanopore sequencing has been used extensively outside the laboratory in many different environments. Researchers have taken the MinION to the Arctic for characterizing permafrost ice wedge microbial communities (47), to the military training grounds at NATO’s Counter Terrorism and Technology Centre in Alberta, Canada, for detection of biological agents (48), and even on the International Space Station to determine how the sequencer performs in space using viral, bacterial, and mouse DNA (49). The MinION has also been used in West Africa during an ongoing Ebola epidemic, where having genome sequencing of the virus on-site allowed for real-time surveillance of the outbreak (50). In these types of situations where resources are scarce and a virus such as Ebola virus can rapidly evolve, having nanopore sequencing available for genomic surveillance can greatly aid in pathogen identification and in monitoring patient responses to vaccines and treatments.
The MinION platform has also been used for research in a number of clinically relevant infectious disease applications. Bacterial and fungal identification of clinical isolates using 16S rRNA and ITS gene sequencing has been successful (31). Researchers were able to obtain 99% accuracy of the 16S rRNA and ITS genes using the 9.5 version flow cells, making the option of targeted sequencing feasible for clinical laboratories on the MinION. Nanopore sequencing has also been used to successfully identify pathogens in clinical cases where identification of the infectious agent can be difficult (51, 52). mWGS with nanopore sequencing was carried out on seven DNA isolates from resected heart valve tissue obtained from patients diagnosed with infective endocarditis (51). Although all seven samples were determined to be culture negative by traditional microbiology testing, species identification of pathogens, which included Streptococcus, Coxiella, and Bartonella spp., was attained in all cases. Prosthetic joint infections, similar to endocarditis, can also be difficult to properly diagnose and treat (53). In a recent study, nine samples, seven culture positive and two culture negative, obtained from sonication fluid were sequenced by mWGS on both the Illumina and ONT platforms (52). Results from the ONT platform corresponded with metagenomics Illumina MiSeq sequencing and culture-based methodologies with the exception that one sample had better species resolution with ONT compared to Illumina sequencing. Furthermore, two samples were found to have an additional species called with the nanopore sequencing pipeline compared to culture-based methods, indicating that ONT sequencing could provide further sensitivity for microorganism detection. Nanopore sequencing can also be applied in cases where a pathogen identified by normal methodologies requires rapid confirmatory testing. Targeted 16S rRNA gene sequencing with a MinION flow cell was performed on a blood-culture isolate obtained from a patient with meningitis (54). Taxonomic analysis confirmed the infectious agent was Campylobacter fetus, a zoonotic pathogen that rarely causes meningitis in humans.
Not only is pathogen identification important in clinical laboratories, but rapid and accurate AST results can significantly impact time to effective antimicrobial therapy. Long-read nanopore sequencing can identify the presence or absence of resistance genes, from which the phenotype of resistance can be inferred. A proof-of-principle study of 40 Klebsiella pneumoniae blood isolates evaluated different data analysis methods for AST prediction, including a real-time analysis approach to identify AMR genes and a nanopore assembly approach (55). That study found 77% agreement with phenotypic methods for a real-time nanopore analysis and 92% agreement with an assembly nanopore approach. Importantly, if employed in real time on a patient isolate, these methods would have hypothetically shortened median time to effective antimicrobial therapy by 20 and 26 h, respectively. In other less clinically oriented studies, extensive characterization of the genomic context of AMR determinants has been elucidated in Escherichia, Salmonella, Klebsiella, and Enterococcus (56–60). Complete plasmid sequences, which are difficult to assemble with short-read platforms, have been resolved using ONT on a host of Enterobacteriaceae species (33, 59). The long reads generated by nanopore sequencing allowed for detection and mapping of mobile AMR elements in a multidrug-resistant strain of enteroaggregative Escherichia coli (57). Despite high error rates, a 2015 study using the earlier iteration of the MinION flow cell looked at AMR in four Enterobacteriaceae species, an Acinetobacter baumannii isolate, and a methicillin-resistant Staphylococcus aureus and found that with even coverage across the genome a specific subset of AMR genes can be accurately called (61). The number of reads necessary to confidently call AMR is not yet well defined. One recent study argues that only one relevant read is necessary to call AMR for a particular drug due to a lower threshold being allowed for long-read sequencing (51), while others have used a threshold of 10 reads (55). In addition to AMR, nanopore sequencing can also provide other genomic data of clinical use. For example, when performing WGS on clinical isolates with both ONT and Illumina sequencing, identification of mutations that may lead to escape of vaccine-induced immunity have been elucidated for Bordetella pertussis (36, 62).
Nanopore sequencing has also been used to identify clinically relevant viruses, often with extensive coverage of entire viral genomes. A 2015 study using an earlier version of the MinION correctly characterized three distinct strains of poxviruses despite an error rate of approximately 30% (63). Another study reported on the detection of chikungunya virus, Ebola virus, and hepatitis C virus from blood samples, with a total turnaround time from sample to report as <6 h (64). Whole genomes of influenza viruses have been sequenced using ONT, demonstrating the use of this technology to track emerging strains of influenza that could potentially be utilized for better vaccine preparation (65).
NEXT STEPS FOR NANOPORE SEQUENCING IN THE CLINICAL LABORATORY
Although application of nanopore technology in the clinical laboratory is still in the research phase, there is significant potential for its use in personalized medicine. The size of the sequencer and cost for utilization makes it ideal for any size clinical laboratory, with the main caveat that appropriately trained personnel are available to carry out method validation, perform nucleic acid extraction, carry out library preparation and sequencing, and analyze the data. Not only would nanopore sequencing be useful for characterizing microorganisms for infectious disease diagnostics, it could be a useful tool for monitoring the human microbiome. The field of microbiome research has exploded over the past 10 years and may become important in clinical medicine in the future (66, 67). For example, the gut microbiome could be monitored over time in individuals undergoing repeated or long-term antibiotic therapy, which can lead to dysbiosis in the gut and other gastrointestinal disorders (68, 69). Nanopore sequencing may also be helpful in characterizing the lung microbiome of individual patients with cystic fibrosis and then diagnosing infection during a pulmonary exacerbation. These are just a few examples of how nanopore sequencing can be utilized in medicine, and the possibilities extend far further than what is mentioned here.
CONCLUSIONS
NGS proof-of-principle assays have been widely used in research to identify and characterize infectious disease agents. Numerous challenges and unknowns limit many clinical laboratories from implementing this technology in-house. More outcomes research, as well as the use of standardized databases, appropriate proficiency materials, and validated thresholds for reporting detected pathogens, will be of utmost importance. The correlations between AMR genotype and phenotype are still not completely understood, which can lead to difficulties in data interpretation. In addition, training and maintaining staff with the appropriate expertise may be a major hurdle for some diagnostic laboratories. It is unlikely to completely replace any current method of conventional diagnostic testing in the near future. Despite these challenges for implementation, mWGS is a promising tool that could provide clinically relevant information such as universal pathogen detection and AMR prediction in a single assay.
Although many platforms for NGS exist, third-generation nanopore sequencing offers many solutions to the current problems of using mWGS for infectious disease diagnostics. It has been successfully utilized for pathogen detection, AMR prediction, and characterization of mixed microbial communities. As improvements continue to be made toward higher accuracy and robust performance, the clear advantages in cost, turnaround time, and user-friendly bioinformatics will likely make it a viable option in the near future for clinical laboratories wanting to implement NGS in-house for infectious disease detection and mWGS.
ACKNOWLEDGMENTS
We thank Stephen Paige for his input and review of the manuscript.
We declare that we have no conflicts of interest.
REFERENCES
- 1.Goldberg B, Sichtig H, Geyer C, Ledeboer N, Weinstock GM. 2015. Making the leap from research laboratory to clinic: challenges and opportunities for next-generation sequencing in infectious disease diagnostics. mBio 6:e01888. doi: 10.1128/mBio.01888-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Forbes JD, Knox NC, Ronholm J, Pagotto F, Reimer A. 2017. Metagenomics: the next culture-independent game changer. Front Microbiol 8:1069. doi: 10.3389/fmicb.2017.01069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Shendure J, Ji H. 2008. Next-generation DNA sequencing. Nat Biotechnol 26:1135–1145. doi: 10.1038/nbt1486. [DOI] [PubMed] [Google Scholar]
- 4.Gu W, Miller S, Chiu CY. 2019. Clinical metagenomic next-generation sequencing for pathogen detection. Annu Rev Pathol 14:319–338. doi: 10.1146/annurev-pathmechdis-012418-012751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Nagahashi M, Shimada Y, Ichikawa H, Kameyama H, Takabe K, Okuda S, Wakai T. 2019. Next generation sequencing-based gene panel tests for the management of solid tumors. Cancer Sci 110:6–15. doi: 10.1111/cas.13837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hirsch B, Endris V, Lassmann S, Weichert W, Pfarr N, Schirmacher P, Kovaleva V, Werner M, Bonzheim I, Fend F, Sperveslage J, Kaulich K, Zacher A, Reifenberger G, Köhrer K, Stepanow S, Lerke S, Mayr T, Aust DE, Baretton G, Weidner S, Jung A, Kirchner T, Hansmann ML, Burbat L, von der Wall E, Dietel M, Hummel M. 2018. Multicenter validation of cancer gene panel-based next-generation sequencing for translational research and molecular diagnostics. Virchows Arch 472:557–565. doi: 10.1007/s00428-017-2288-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lefterova MI, Suarez CJ, Banaei N, Pinsky BA. 2015. Next-generation sequencing for infectious disease diagnosis and management a report of the association for molecular pathology. J Mol Diagn 17:623–634. doi: 10.1016/j.jmoldx.2015.07.004. [DOI] [PubMed] [Google Scholar]
- 8.Deshmukh D, Joseph J, Chakrabarti M, Sharma S, Jayasudha R, Sama KC, Sontam B, Tyagi M, Narayanan R, Shivaji S. 2019. New insights into culture negative endophthalmitis by unbiased next generation sequencing. Sci Rep 9:844. doi: 10.1038/s41598-018-37502-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Su M, Satola SW, Read TD. 2018. Genome-based prediction of bacterial antibiotic resistance. J Clin Microbiol 57:e01405-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Schlaberg R, Chiu CY, Miller S, Procop GW, Weinstock G. 2017. Validation of metagenomic next-generation sequencing tests for universal pathogen detection. Arch Pathol Lab Med 141:776–786. doi: 10.5858/arpa.2016-0539-RA. [DOI] [PubMed] [Google Scholar]
- 11.Miller S, Naccache SN, Samayoa E, Messacar K, Arevalo S, Federman S, Stryke D, Pham E, Fung B, Bolosky WJ, Ingebrigtsen D, Lorizio W, Paff SM, Leake JA, Pesano R, DeBiasi R, Dominguez S, Chiu CY. 2019. Laboratory validation of a clinical metagenomic sequencing assay for pathogen detection in cerebrospinal fluid. Genome Res 29:831–842. doi: 10.1101/gr.238170.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Wilson MR, Sample HA, Zorn KC, Arevalo S, Yu G, Neuhaus J, Federman S, Stryke D, Briggs B, Langelier C, Berger A, Douglas V, Josephson SA, Chow FC, Fulton BD, DeRisi JL, Gelfand JM, Naccache SN, Bender J, Dien Bard J, Murkey J, Carlson M, Vespa PM, Vijayan T, Allyn PR, Campeau S, Humphries RM, Klausner JD, Ganzon CD, Memar F, Ocampo NA, Zimmermann LL, Cohen SH, Polage CR, DeBiasi RL, Haller B, Dallas R, Maron G, Hayden R, Messacar K, Dominguez SR, Miller S, Chiu CY. 2019. Clinical metagenomic sequencing for diagnosis of meningitis and encephalitis. N Engl J Med 380:2327–2340. doi: 10.1056/NEJMoa1803396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Charalampous T, Richardson H, Kay GL, Baldan R, Jeanes C, Rae D, Grundy S, Turner DJ, Wain J, Leggett RM, Livermore DM, O’Grady J. 2018. Rapid diagnosis of lower respiratory infection using nanopore-based clinical metagenomics. bioRxiv 387548. [DOI] [PubMed] [Google Scholar]
- 14.Couto N, Schuele L, Raangs EC, Machado MP, Mendes CI, Jesus TF, Chlebowicz M, Rosema S, Ramirez M, Carriço JA, Autenrieth IB, Friedrich AW, Peter S, Rossen JW. 2018. Critical steps in clinical shotgun metagenomics for the concomitant detection and typing of microbial pathogens. Sci Rep 8:13767. doi: 10.1038/s41598-018-31873-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Petti CA, Polage CR, Schreckenberger P. 2005. The role of 16S rRNA gene sequencing in identification of microorganisms misidentified by conventional methods. J Clin Microbiol 43:6123–6125. doi: 10.1128/JCM.43.12.6123-6125.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Mignard S, Flandrois JP. 2006. 16S rRNA sequencing in routine bacterial identification: a 30-month experiment. J Microbiol Methods 67:574–581. doi: 10.1016/j.mimet.2006.05.009. [DOI] [PubMed] [Google Scholar]
- 17.Janda JM, Abbott SL. 2007. 16S rRNA gene sequencing for bacterial identification in the diagnostic laboratory: pluses, perils, and pitfalls. J Clin Microbiol 45:2761–2764. doi: 10.1128/JCM.01228-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kim M, Oh H, Park S, Chun J. 2014. Towards a taxonomic coherence between average nucleotide identity and 16S rRNA gene sequence similarity for species demarcation of prokaryotes. Int J Syst Evol Microbiol 64:346–351. doi: 10.1099/ijs.0.059774-0. [DOI] [PubMed] [Google Scholar]
- 19.Schoch CL, Seifert KA, Huhndorf S, Robert V, Spouge JL, Levesque CA, Chen W, Consortium FB. 2012. Nuclear ribosomal internal transcribed spacer (ITS) region as a universal DNA barcode marker for Fungi. Proc Natl Acad Sci U S A 109:6241–6246. doi: 10.1073/pnas.1117018109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Frank JA, Reich CI, Sharma S, Weisbaum JS, Wilson BA, Olsen GJ. 2008. Critical evaluation of two primers commonly used for amplification of bacterial 16S rRNA genes. Appl Environ Microbiol 74:2461–2470. doi: 10.1128/AEM.02272-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Simner PJ, Miller HB, Breitwieser FP, Pinilla Monsalve G, Pardo CA, Salzberg SL, Sears CL, Thomas DL, Eberhart CG, Carroll KC. 2018. Development and optimization of metagenomic next-generation sequencing methods for cerebrospinal fluid diagnostics. J Clin Microbiol 56:e00472-18. doi: 10.1128/JCM.00472-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Sichtig H, Minogue T, Yan Y, Stefan C, Hall A, Tallon L, Sadzewicz L, Nadendla S, Klimke W, Hatcher E, Shumway M, Aldea DL, Allen J, Koehler J, Slezak T, Lovell S, Schoepp R, Scherf U. 2019. FDA-ARGOS is a database with public quality-controlled reference genomes for diagnostic use and regulatory science. Nat Commun 10:3313. doi: 10.1038/s41467-019-11306-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Deurenberg RH, Bathoorn E, Chlebowicz MA, Couto N, Ferdous M, García-Cobos S, Kooistra-Smid AMD, Raangs EC, Rosema S, Veloo ACM, Zhou K, Friedrich AW, Rossen J. 2017. Application of next generation sequencing in clinical microbiology and infection prevention. J Biotechnol 243:16–24. doi: 10.1016/j.jbiotec.2016.12.022. [DOI] [PubMed] [Google Scholar]
- 24.Rohit A, Suresh Kumar D, Dhinakaran I, Joy J, Vijay Kumar D, Kumar Ballamoole K, Karunasagar I, Karola P, Dag H. 2019. Whole-genome-based analysis reveals multiclone Serratia marcescens outbreaks in a non-neonatal intensive care unit setting in a tertiary care hospital in India. J Med Microbiol 68:616–621. doi: 10.1099/jmm.0.000947. [DOI] [PubMed] [Google Scholar]
- 25.Roy S, Hartley J, Dunn H, Williams R, Williams C, Breuer J. 2019. Whole-genome sequencing provides data for stratifying infection prevention and control management of nosocomial influenza A. Clin Infect Dis 2019:ciz020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Costa DM, Johani K, Melo DS, Lopes LKO, Lopes Lima LKO, Tipple AFV, Hu H, Vickery K. 2019. Biofilm contamination of high-touched surfaces in intensive care units: epidemiology and potential impacts. Lett Appl Microbiol 68:269–276. doi: 10.1111/lam.13127. [DOI] [PubMed] [Google Scholar]
- 27.Eichenberger EM, Thaden JT. 2019. Epidemiology and mechanisms of resistance of extensively drug resistant Gram-negative bacteria. Antibiotics 8:37–21. doi: 10.3390/antibiotics8020037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Laver T, Harrison J, O’Neill PA, Moore K, Farbos A, Paszkiewicz K, Studholme DJ. 2015. Assessing the performance of the Oxford Nanopore Technologies MinION. Biomol Detect Quantif 3:1–8. doi: 10.1016/j.bdq.2015.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Payne A, Holmes N, Rakyan V, Loose M. 2019. BulkVis: a graphical viewer for Oxford nanopore bulk FAST5 files. Bioinformatics 35:2193–2198. doi: 10.1093/bioinformatics/bty841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Lu H, Giordano F, Ning Z. 2016. Oxford Nanopore MinION sequencing and genome assembly. Genomics Proteomics Bioinformatics 14:265–279. doi: 10.1016/j.gpb.2016.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ashikawa S, Tarumoto N, Imai K, Sakai J, Kodana M, Kawamura T, Ikebuchi K, Murakami T, Mitsutake K, Maesaki S, Maeda T. 2018. Rapid identification of pathogens from positive blood culture bottles with the MinION nanopore sequencer. J Med Microbiol 67:1589–1595. doi: 10.1099/jmm.0.000855. [DOI] [PubMed] [Google Scholar]
- 32.Wick RR, Judd LM, Gorrie CL, Holt KE. 2017. Completing bacterial genome assemblies with multiplex MinION sequencing. Microb Genom 3:e000132. doi: 10.1099/mgen.0.000132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.George S, Pankhurst L, Hubbard A, Votintseva A, Stoesser N, Sheppard AE, Mathers A, Norris R, Navickaite I, Eaton C, Iqbal Z, Crook DW, Phan H. 2017. Resolving plasmid structures in Enterobacteriaceae using the MinION nanopore sequencer: assessment of MinION and MinION/Illumina hybrid data assembly approaches. Microb Genom 3:e000118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Judge K, Hunt M, Reuter S, Tracey A, Quail MA, Parkhill J, Peacock SJ. 2016. Comparison of bacterial genome assembly software for MinION data and their applicability to medical microbiology. Microb Genom 2:e000085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Tyson JR, O’Neil NJ, Jain M, Olsen HE, Hieter P, Snutch TP. 2018. MinION-based long-read sequencing and assembly extends the Caenorhabditis elegans reference genome. Genome Res 28:266–274. doi: 10.1101/gr.221184.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bouchez V, Baines SL, Guilot S, Brisse S. 2018. Complete genome sequences of Bordetella pertussis clinical isolate FR5810 and reference strain Tohama from combined Oxford Nanopore and Illumina sequencing. Gen Seq 7:e01207-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Luo R, Zimin A, Workman R, Fan Y, Pertea G, Grossman N, Wear MP, Jia B, Miller H, Casadevall A, Timp W, Zhang SX, Salzberg SL. 2017. First draft genome sequence of the pathogenic fungus Lomentospora prolificans (formerly Scedosporium prolificans). G3 (Bethesda) 7:3831–3836. doi: 10.1534/g3.117.300107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Wick RR, Judd LM, Holt K. 2019. Performance of neural network basecalling tools for Oxford Nanopore sequencing. bioRxiv 543439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Karst SM, Ziels RM, Kirkegaard RH, Albertsen M. 2019. Enabling high-accuracy long-read amplicon sequences using unique molecular identifiers and Nanopore sequencing. bioRxiv 645903. [DOI] [PubMed] [Google Scholar]
- 40.Kono N, Arakawa K. 2019. Nanopore sequencing: review of potential applications in functional genomics. Dev Growth Differ 61:316–326. doi: 10.1111/dgd.12608. [DOI] [PubMed] [Google Scholar]
- 41.Kim H-S, Jeon S, Kim C, Kim YK, Cho YS, Kim J, Blazyte A, Manica A, Lee S, Bhak J. 2019. Chromosome-scale assembly comparison of the Korean Reference Genome KOREF from PromethION and PacBio with Hi-C mapping information. bioRxiv 674804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Jain M, Koren S, Miga KH, Quick J, Rand AC, Sasani TA, Tyson JR, Beggs AD, Dilthey AT, Fiddes IT, Malla S, Marriott H, Nieto T, O’Grady J, Olsen HE, Pedersen BS, Rhie A, Richardson H, Quinlan AR, Snutch TP, Tee L, Paten B, Phillippy AM, Simpson JT, Loman NJ, Loose M. 2018. Nanopore sequencing and assembly of a human genome with ultra-long reads. Nat Biotechnol 36:338–345. doi: 10.1038/nbt.4060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Minot SS, Krumm N, Greenfield NB. 2015. One Codex: a sensitive and accurate data platform for genomic microbial identification. bioRxiv 27607. [Google Scholar]
- 44.Santiago-Rodriguez TM, Fornaciari A, Fornaciari G, Luciani S, Marota I, Vercellotti G, Toranzos GA, Giuffra V, Cano RJ. 2019. Commensal and pathogenic members of the dental calculus microbiome of Badia Pozzeveri individuals from the 11th to 19th centuries. Genes 10:299. doi: 10.3390/genes10040299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kalan LR, Meisel JS, Loesche MA, Horwinski J, Soaita I, Chen X, Uberoi A, Gardner SE, Grice EA. 2019. Strain- and species-level variation in the microbiome of diabetic wounds is associated with clinical outcomes and therapeutic efficacy. Cell Host Microbe 25:641–655. doi: 10.1016/j.chom.2019.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Petersen LM, Bautista EJ, Nguyen H, Hanson BM, Chen L, Lek SH, Sodergren E, Weinstock GM. 2017. Community characteristics of the gut microbiomes of competitive cyclists. Microbiome 5:98. doi: 10.1186/s40168-017-0320-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Goordial J, Altshuler I, Hindson K, Chan-Yam K, Marcolefas E, Whyte LG. 2017. In situ field sequencing and life detection in remote (79°26′N) Canadian high arctic permafrost ice wedge microbial communities. Front Microbiol 8:2594. doi: 10.3389/fmicb.2017.02594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Walter MC, Zwirglmaier K, Vette P, Holowachuk SA, Stoecker K, Genzel GH, Antwerpen MH. 2017. MinION as part of a biomedical rapidly deployable laboratory. J Biotechnol 250:16–22. doi: 10.1016/j.jbiotec.2016.12.006. [DOI] [PubMed] [Google Scholar]
- 49.Castro-Wallace SL, Chiu CY, John KK, Stahl SE, Rubins KH, McIntyre ABR, Dworkin JP, Lupisella ML, Smith DJ, Botkin DJ, Stephenson TA, Juul S, Turner DJ, Izquierdo F, Federman S, Stryke D, Somasekar S, Alexander N, Yu G, Mason CE, Burton AS. 2017. Nanopore DNA sequencing and genome assembly on the international space station. Sci Rep 7:18022. doi: 10.1038/s41598-017-18364-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Quick J, Loman NJ, Duraffour S, Simpson JT, Severi E, et al. . 2016. Real-time, portable genome sequencing for Ebola surveillance. Nature 530:228–232. doi: 10.1038/nature16996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Cheng J, Hu H, Kang Y, Chen W, Fang W, Wang K, Zhang Q, Fu A, Zhou S, Cheng C, Cao Q, Wang F, Lee S, Zhou Z. 2018. Identification of pathogens in culture-negative infective endocarditis cases by metagenomic analysis. Ann Clin Microbiol Antimicrob 17:1–11. doi: 10.1186/s12941-018-0294-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Sanderson ND, Street TL, Foster D, Swann J, Atkins BL, Brent AJ, McNally MA, Oakley S, Taylor A, Peto TEA, Crook DW, Eyre DW. 2018. Real-time analysis of nanopore-based metagenomic sequencing from infected orthopaedic devices. BMC Genomics 19:714. doi: 10.1186/s12864-018-5094-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Osmon DR. 2017. Microbiology and antimicrobial challenges of prosthetic joint infection. J Am Acad Orthop Surg 25:S17–S19. doi: 10.5435/JAAOS-D-16-00639. [DOI] [PubMed] [Google Scholar]
- 54.Moon J, Kim N, Lee HS, Shin HR, Lee ST, Jung KH, Park KIl, Lee SK, Chu K. 2017. Campylobacter fetus meningitis confirmed by a 16S rRNA gene analysis using the MinION nanopore sequencer, South Korea, 2016. Emerg Microbes Infect 6:e94. doi: 10.1038/emi.2017.81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Tamma PD, Fan Y, Bergman Y, Pertea G, Kazmi AQ, Lewis S, Carroll KC, Schatz MC, Timp W, Simner PJ. 2018. Applying rapid whole-genome sequencing to predict phenotypic antimicrobial susceptibility testing results among carbapenem-resistant Klebsiella pneumoniae clinical isolates. Antimicrob Agents Chemother 63:e01923-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Long H, Feng Y, Ma K, Liu L, McNally A, Zong Z. 2019. The cotransfer of plasmid-borne colistin-resistant genes mcr-1 and mcr-3.5, the carbapenemase gene blaNDM-5 and the 16S methylase gene rmtB from Escherichia coli. Sci Rep 9:696. doi: 10.1038/s41598-018-37125-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Greig DR, Dallman TJ, Hopkins KL, Jenkins C. 2018. MinION nanopore sequencing identifies the position and structure of bacterial antibiotic resistance determinants in a multidrug-resistant strain of enteroaggregative Escherichia coli. Microb Genom 4:e000213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Li R, Chen K, Wai Chi Chan E, Chen S. 2018. Resolution of dynamic MDR structures among the plasmidome of Salmonella using MinION single-molecule, long-read sequencing. J Antimicrob Chemother 73:2691–2695. doi: 10.1093/jac/dky243. [DOI] [PubMed] [Google Scholar]
- 59.Lemon JK, Khil PP, Frank KM, Dekker JP. 2017. Rapid nanopore sequencing of plasmids and resistance gene detection in clinical isolates. J Clin Microbiol 55:3530–3543. doi: 10.1128/JCM.01069-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hansen TA, Pedersen MS, Nielsen LG, Ma CMG, Søes LM, Worning P, Østergaard C, Westh H, Pinholt M, Schønning K. 2018. Emergence of a vancomycin-variable Enterococcus faecium ST1421 strain containing a deletion in vanX. J Antimicrob Chemother 73:2936–2940. doi: 10.1093/jac/dky308. [DOI] [PubMed] [Google Scholar]
- 61.Judge K, Harris SR, Reuter S, Parkhill J, Peacock SJ. 2015. Early insights into the potential of the Oxford Nanopore MinION for the detection of antimicrobial resistance genes. J Antimicrob Chemother 70:2775–2778. doi: 10.1093/jac/dkv206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Parajuli P, Deimel LP, Verma NK. 2019. Genome analysis of Shigella flexneri serotype 3b strain SFL1520 reveals significant horizontal gene acquisitions including a multidrug resistance cassette. Genome Biol Evol 11:776–785. doi: 10.1093/gbe/evz026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kilianski A, Haas JL, Corriveau EJ, Liem AT, Willis KL, Kadavy DR, Rosenzweig CN, Minot SS. 2015. Bacterial and viral identification and differentiation by amplicon sequencing on the MinION nanopore sequencer. Gigascience 4:1–8. doi: 10.1186/s13742-015-0051-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Greninger AL, Naccache SN, Federman S, Yu G, Mbala P, Bres V, Stryke D, Bouquet J, Somasekar S, Linnen JM, Dodd R, Mulembakani P, Schneider BS, Muyembe-Tamfum JJ, Stramer SL, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Imai K, Tamura K, Tanigaki T, Takizawa M, Nakayama E, Taniguchi T, Okamoto M, Nishiyama Y, Tarumoto N, Mitsutake K, Murakami T, Maesaki S, Maeda T. 2018. Whole genome sequencing of influenza A and B viruses with the MinION sequencer in the clinical setting: a pilot study. Front Microbiol 9:2748. doi: 10.3389/fmicb.2018.02748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Kuntz TM, Gilbert JA. 2017. Introducing the microbiome into precision medicine. Trends Pharmacol Sci 38:81–91. doi: 10.1016/j.tips.2016.10.001. [DOI] [PubMed] [Google Scholar]
- 67.Relman DA. 2015. The human microbiome and the future practice of medicine. JAMA 314:1127–1128. doi: 10.1001/jama.2015.10700. [DOI] [PubMed] [Google Scholar]
- 68.Francino MP. 2016. Antibiotics and the human gut microbiome: dysbioses and accumulation of resistances. Front Microbiol 6:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Dethlefsen L, 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 108:4554–4561. doi: 10.1073/pnas.1000087107. [DOI] [PMC free article] [PubMed] [Google Scholar]