52
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Translating RNA sequencing into clinical diagnostics: opportunities and challenges

      review-article

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Key Points

          • RNA-based measurements have the potential for application across diverse areas of human health, including disease diagnosis, prognosis and therapeutic selection. Current clinical applications include infectious diseases, cancer, transplant medicine and fetal monitoring.

          • RNA sequencing (RNA-seq) allows for the detection of a wide variety of RNA species, including mRNA, non-coding RNA, pathogen RNA, chimeric gene fusions, transcript isoforms and splice variants, and provides the capability to quantify known, pre-defined RNA species and rare RNA transcript variants within a sample. In addition to differential expression and detection of novel transcripts, RNA-seq also supports the detection of mutations and germline variation for hundreds to thousands of expressed genetic variants, facilitating assessment of allele-specific expression of these variants.

          • Circulating RNAs and small regulatory RNAs, such as microRNAs, are very stable. These RNA species are vigorously being tested for their potential as biomarkers. However, there are currently few agreed upon methods for isolation or quantitative measurements and a current lack of quality controls that can be used to test platform accuracy and sample preparation quality.

          • Analytical, bioinformatic and regulatory challenges exist, and ongoing efforts toward the establishment of benchmark standards, assay optimization for clinical conditions and demonstration of assay reproducibility are required to expand the clinical utility of RNA-seq.

          Supplementary information

          The online version of this article (doi:10.1038/nrg.2016.10) contains supplementary material, which is available to authorized users.

          Abstract

          RNA sequencing (RNA-seq) is a powerful approach for comprehensive analyses of transcriptomes. This Review describes the widespread potential applications of RNA-seq in clinical medicine, such as detecting disease-associated mutations and gene expression disruptions, as well as characteristic non-coding RNAs, circulating extracellular RNAs or pathogen RNAs. The authors also highlight the challenges in adopting RNA-seq routinely into clinical practice.

          Supplementary information

          The online version of this article (doi:10.1038/nrg.2016.10) contains supplementary material, which is available to authorized users.

          Abstract

          With the emergence of RNA sequencing (RNA-seq) technologies, RNA-based biomolecules hold expanded promise for their diagnostic, prognostic and therapeutic applicability in various diseases, including cancers and infectious diseases. Detection of gene fusions and differential expression of known disease-causing transcripts by RNA-seq represent some of the most immediate opportunities. However, it is the diversity of RNA species detected through RNA-seq that holds new promise for the multi-faceted clinical applicability of RNA-based measures, including the potential of extracellular RNAs as non-invasive diagnostic indicators of disease. Ongoing efforts towards the establishment of benchmark standards, assay optimization for clinical conditions and demonstration of assay reproducibility are required to expand the clinical utility of RNA-seq.

          Supplementary information

          The online version of this article (doi:10.1038/nrg.2016.10) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references95

          • Record: found
          • Abstract: found
          • Article: not found

          Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer.

          The 21-gene recurrence score (RS) assay quantifies the likelihood of distant recurrence in women with estrogen receptor-positive, lymph node-negative breast cancer treated with adjuvant tamoxifen. The relationship between the RS and chemotherapy benefit is not known. The RS was measured in tumors from the tamoxifen-treated and tamoxifen plus chemotherapy-treated patients in the National Surgical Adjuvant Breast and Bowel Project (NSABP) B20 trial. Cox proportional hazards models were utilized to test for interaction between chemotherapy treatment and the RS. A total of 651 patients were assessable (227 randomly assigned to tamoxifen and 424 randomly assigned to tamoxifen plus chemotherapy). The test for interaction between chemotherapy treatment and RS was statistically significant (P = .038). Patients with high-RS (> or = 31) tumors (ie, high risk of recurrence) had a large benefit from chemotherapy (relative risk, 0.26; 95% CI, 0.13 to 0.53; absolute decrease in 10-year distant recurrence rate: mean, 27.6%; SE, 8.0%). Patients with low-RS (< 18) tumors derived minimal, if any, benefit from chemotherapy treatment (relative risk, 1.31; 95% CI, 0.46 to 3.78; absolute decrease in distant recurrence rate at 10 years: mean, -1.1%; SE, 2.2%). Patients with intermediate-RS tumors did not appear to have a large benefit, but the uncertainty in the estimate can not exclude a clinically important benefit. The RS assay not only quantifies the likelihood of breast cancer recurrence in women with node-negative, estrogen receptor-positive breast cancer, but also predicts the magnitude of chemotherapy benefit.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Tailoring therapies—improving the management of early breast cancer: St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2015

            The 14th St Gallen International Breast Cancer Conference (2015) reviewed new evidence on locoregional and systemic therapies for early breast cancer. This manuscript presents news and progress since the 2013 meeting, provides expert opinion on almost 200 questions posed to Consensus Panel members, and summarizes treatment-oriented classification of subgroups and treatment recommendations.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Computational methods for transcriptome annotation and quantification using RNA-seq.

              High-throughput RNA sequencing (RNA-seq) promises a comprehensive picture of the transcriptome, allowing for the complete annotation and quantification of all genes and their isoforms across samples. Realizing this promise requires increasingly complex computational methods. These computational challenges fall into three main categories: (i) read mapping, (ii) transcriptome reconstruction and (iii) expression quantification. Here we explain the major conceptual and practical challenges, and the general classes of solutions for each category. Finally, we highlight the interdependence between these categories and discuss the benefits for different biological applications.
                Bookmark

                Author and article information

                Contributors
                dcraig@tgen.org
                Journal
                Nat Rev Genet
                Nat. Rev. Genet
                Nature Reviews. Genetics
                Nature Publishing Group UK (London )
                1471-0056
                1471-0064
                21 March 2016
                2016
                : 17
                : 5
                : 257-271
                Affiliations
                [1 ]GRID grid.250942.8, ISNI 0000 0004 0507 3225, Center for Translational Innovation, Translational Genomics Research Institute, ; Phoenix, 85004 Arizona USA
                [2 ]GRID grid.250942.8, ISNI 0000 0004 0507 3225, Neurogenomics Division, , Translational Genomics Research Institute, ; Phoenix, 85004 Arizona USA
                [3 ]GRID grid.250942.8, ISNI 0000 0004 0507 3225, Pathogen Genomics Division, , Translational Genomics Research Institute, ; Flagstaff, 86001 Arizona USA
                [4 ]GRID grid.250942.8, ISNI 0000 0004 0507 3225, Integrated Cancer Genomics Division, , Translational Genomics Research Institute, ; Phoenix, 85004 Arizona USA
                Article
                BFnrg201610
                10.1038/nrg.2016.10
                7097555
                26996076
                e2e038dd-f070-4e55-866c-226568f05e41
                © Nature Publishing Group, a division of Macmillan Publishers Limited. All Rights Reserved. 2016

                This article is made available via the PMC Open Access Subset for unrestricted research re-use and secondary analysis in any form or by any means with acknowledgement of the original source. These permissions are granted for the duration of the World Health Organization (WHO) declaration of COVID-19 as a global pandemic.

                History
                Categories
                Article
                Custom metadata
                © Springer Nature Limited 2016

                rna sequencing,next-generation sequencing,gene expression profiling,transcriptomics,genome-wide analysis of gene expression,cancer genetics,pathogens,non-coding rnas,diagnostic markers

                Comments

                Comment on this article