Omics Technologies in Clinical Practice:
A Practical Review for Physicians
Abstract
The rapid advancement of high-throughput molecular technologies has ushered in the era of "omics" in medicine, transforming our understanding of disease pathophysiology and approach to patient care. This review provides a practical overview of the major omics technologies—genomics, transcriptomics, proteomics, metabolomics, and emerging multi-omics approaches—with emphasis on their current clinical applications and integration into medical practice. We discuss practical considerations for physicians, including test selection, result interpretation, implementation challenges, and emerging frameworks for clinical decision support. Case examples across specialties illustrate how omics technologies can enhance diagnosis, guide targeted therapies, enable risk stratification, and facilitate personalized treatment approaches. While acknowledging limitations and barriers to widespread adoption, this review offers a roadmap for physicians to effectively navigate and utilize these powerful technologies to improve clinical outcomes.
Keywords: Omics, precision medicine, genomics, transcriptomics, proteomics, metabolomics, clinical implementation, personalized medicine
Introduction
The suffix "-omics" denotes the comprehensive assessment of a set of molecules, with each omics field examining a different layer of biological information. Collectively, omics technologies enable an unprecedented systems-level understanding of human biology in health and disease.^1^ These technologies have evolved from primarily research tools to increasingly practical clinical applications, driven by reduced costs, improved analytical methods, and accumulating evidence of clinical utility.^2^
For many physicians, however, the rapidly evolving omics landscape presents challenges in understanding which technologies are clinically mature, how to appropriately order and interpret tests, and how to integrate complex molecular data into clinical decision-making.^3^ This review aims to bridge this knowledge gap by providing a practical overview of major omics technologies as they relate to clinical practice, focusing on applications that have demonstrated clinical utility or are on the cusp of clinical implementation.
Overview of Major Omics Technologies
Genomics
Genomics—the study of an organism's complete DNA sequence—has made the greatest inroads into clinical practice among omics technologies.^4^ Clinical genomic testing ranges from targeted genotyping of specific variants to whole exome sequencing (WES) and whole genome sequencing (WGS).
Clinical Applications:
-
Rare Disease Diagnosis: WES and WGS have revolutionized the diagnosis of rare genetic disorders, with diagnostic yields of 25-50% in previously undiagnosed cases.^5^ For example, the Undiagnosed Diseases Network reported a 35% diagnostic rate using genomic sequencing in patients who had remained undiagnosed despite extensive prior evaluation.^6^
-
Pharmacogenomics: Genetic variants affecting drug metabolism (e.g., CYP2C19 for clopidogrel, CYP2D6 for tamoxifen) can guide medication selection and dosing. The Clinical Pharmacogenetics Implementation Consortium (CPIC) provides evidence-based guidelines for incorporating pharmacogenomic data into prescribing decisions for over 25 drug-gene pairs.^7^
-
Cancer Management:
- Germline testing can identify heritable cancer predisposition syndromes (e.g., BRCA1/2, Lynch syndrome) guiding preventive interventions.^8^
- Tumor sequencing identifies actionable mutations to guide targeted therapy selection. The NCI-MATCH and ASCO's TAPUR studies have demonstrated the feasibility and potential benefits of this approach.^9,10^
-
Carrier Screening: Expanded carrier screening can identify reproductive risks for hundreds of recessive and X-linked conditions.^11^
-
Infectious Disease Diagnostics: Metagenomic next-generation sequencing enables comprehensive pathogen detection, particularly valuable in challenging cases like encephalitis, where conventional testing is often negative.^12^
Transcriptomics
Transcriptomics examines the complete set of RNA transcripts produced by the genome under specific circumstances, providing a dynamic picture of gene expression.^13^ Technologies include microarrays and RNA sequencing (RNA-seq).
Clinical Applications:
-
Cancer Classification and Prognostication: Transcriptome-based tests like Oncotype DX (breast cancer), Decipher (prostate cancer), and Allosure (transplant rejection risk) stratify patients to guide treatment decisions.^14^ For example, Oncotype DX generates a recurrence score that predicts chemotherapy benefit in early-stage breast cancer, allowing many women to safely avoid chemotherapy.^15^
-
Infectious Disease Diagnostics: Host transcriptional signatures can distinguish bacterial from viral infections, potentially reducing unnecessary antibiotic use. Tests like HostDx Sepsis are being developed to address this need.^16^
-
Transplant Medicine: RNA expression profiles in blood or allograft biopsies can predict or diagnose rejection, sometimes before clinical manifestations appear.^17^
-
Neuropsychiatric Disorders: Though still emerging, transcriptomic signatures may aid in diagnosing and subtyping conditions like autism spectrum disorder and major depression, potentially guiding treatment selection.^18^
Proteomics
Proteomics studies the entire set of proteins expressed in a cell, tissue, or organism, including post-translational modifications that affect protein function.^19^ Mass spectrometry is the primary technology used for clinical proteomics.
Clinical Applications:
-
Biomarker Discovery and Validation: Proteomic approaches have identified novel biomarkers across numerous diseases. Clinical examples include:
- The Vectra DA test, measuring 12 proteins to assess rheumatoid arthritis disease activity^20^
- The SomaSignal tests, which use the SomaScan® platform to measure thousands of proteins for multiple clinical applications including cardiovascular risk assessment^21^
-
Cancer Diagnostics and Monitoring: Mass spectrometry-based methods can identify cancer-specific protein signatures in blood or other body fluids. For example, OVA1 integrates five protein biomarkers to assess malignancy risk in ovarian masses.^22^
-
Toxicology: Proteomics can identify exposure to toxins and drugs not detectable by standard toxicology panels.^23^
-
Microbial Identification: MALDI-TOF mass spectrometry rapidly identifies bacterial and fungal species from culture, now standard in many clinical microbiology laboratories.^24^
Metabolomics
Metabolomics examines the complete set of small-molecule metabolites, providing a functional readout of physiological processes.^25^ Technologies include nuclear magnetic resonance (NMR) spectroscopy and mass spectrometry.
Clinical Applications:
-
Inborn Errors of Metabolism: Expanded newborn screening using tandem mass spectrometry detects numerous metabolic disorders, enabling early intervention to prevent intellectual disability and death.^26^
-
Cancer Metabolism: Altered metabolic profiles can aid cancer diagnosis, assess aggressiveness, and identify therapeutic targets. PET imaging with various tracers exploits cancer-specific metabolic patterns for clinical diagnosis and monitoring.^27^
-
Diabetes and Metabolic Disorders: Metabolomic profiles can predict diabetes development before traditional markers become abnormal and identify subtypes of diabetes that may respond differently to therapies.^28^
-
Drug Response Prediction: Metabolomic signatures can predict response to various medications, including antidepressants and antihypertensives.^29^
-
Microbiome-Associated Disorders: Metabolites produced by gut microbiota (e.g., trimethylamine N-oxide in cardiovascular disease) have significant clinical implications.^30^
Emerging Multi-Omics Approaches
Integration of multiple omics data types provides more comprehensive insights than any single approach.^31^ Clinical applications are still emerging but show promise in:
-
Complex Disease Subtyping: Identifying molecularly distinct disease subtypes that respond differently to therapy, as demonstrated in studies of asthma, inflammatory bowel disease, and various cancers.^32^
-
Health Monitoring: Longitudinal multi-omics profiling can detect deviations from an individual's baseline, potentially enabling early disease detection, as shown in the Pioneer 100 and subsequent studies.^33^
-
Treatment Selection: Integrating genomic, transcriptomic, and proteomic data can better predict treatment responses than single omics approaches, particularly in oncology.^34^
Clinical Implementation Considerations
Test Selection and Ordering
When considering omics testing, physicians should:
-
Clarify the Clinical Question: Define what specific information will affect clinical decision-making.^35^
-
Understand Test Characteristics: Consider analytical validity (accuracy), clinical validity (predictive value), and clinical utility (impact on outcomes).^36^
-
Consider Testing Strategies:
- For suspected genetic disorders: Consider targeted testing first if clinical presentation strongly suggests specific genes; proceed to broader testing (panels, exome) if targeted testing is negative or clinical presentation is nonspecific.^37^
- For cancer: Distinguish between germline (heritable) testing and somatic (tumor) testing, which answer different clinical questions.^38^
-
Account for Turnaround Time: Some omics tests provide results in hours (e.g., rapid WGS in critically ill infants), while others may take weeks.^39^
-
Verify Insurance Coverage: Many advanced omics tests have variable coverage, potentially creating access disparities.^40^
Interpretation and Clinical Action
Translating complex omics data into clinical decisions requires:
-
Understanding Test Limitations: All omics technologies have specific limitations and potential blind spots:
- WES cannot reliably detect certain variant types (e.g., structural variations, repeat expansions)
- Transcriptome analysis provides a temporal snapshot that may change based on numerous factors
- Metabolomic profiles can be influenced by medications, diet, and sample handling^41^
-
Interdisciplinary Collaboration: Molecular tumor boards and genomic medicine consult services can help interpret complex results and formulate management plans.^42^
-
Prioritizing Actionable Findings: Focus on findings with established clinical implications while acknowledging areas of uncertainty.^43^
-
Managing Incidental Findings: Have a plan for handling secondary findings unrelated to the primary indication for testing. The ACMG provides recommendations for reporting certain actionable incidental findings from genomic sequencing.^44^
-
Systematic Documentation: Document the indication for testing, results, interpretation, and clinical actions in the medical record to enable appropriate follow-up and family counseling.^45^
Ethical and Practical Challenges
Key considerations include:
-
Informed Consent: Patients should understand the scope of testing, potential for incidental findings, implications for family members, and data privacy considerations.^46^
-
Privacy and Data Security: Comprehensive omics data contains highly sensitive information requiring robust protection.^47^
-
Health Disparities: Ensure equitable access to omics technologies across diverse populations. Current genomic databases overrepresent European ancestries, potentially limiting clinical utility in other populations.^48^
-
Managing Patient Expectations: Clearly communicate both the potential and limitations of omics testing to avoid unrealistic expectations.^49^
-
Cost-Effectiveness: Consider the value proposition of omics testing, including downstream costs and benefits beyond the immediate diagnostic yield.^50^
Case Examples of Clinical Applications
Case 1: Pediatric Neurology
A 4-year-old child presents with developmental regression, seizures, and movement disorder of unclear etiology despite extensive conventional workup. Rapid whole genome sequencing identifies a de novo variant in the FOXG1 gene, confirming FOXG1 syndrome. This diagnosis ends the diagnostic odyssey, enables genetic counseling regarding recurrence risk (low), and guides management of specific complications known to occur in this syndrome.^51^
Case 2: Oncology
A 57-year-old woman with metastatic lung adenocarcinoma undergoes comprehensive genomic profiling revealing an ALK rearrangement. Treatment with the ALK inhibitor alectinib results in dramatic and durable response. Later progression prompts repeat molecular profiling, identifying a resistance mutation (ALK G1202R), guiding switch to lorlatinib with renewed response.^52^
Case 3: Infectious Disease
A 32-year-old previously healthy man presents with encephalitis of unknown etiology despite extensive conventional microbiologic testing. Metagenomic next-generation sequencing of cerebrospinal fluid identifies Listeria monocytogenes RNA, guiding appropriate antibiotic therapy and good neurological recovery.^53^
Case 4: Cardiology
A 45-year-old woman with chest pain undergoes protein-based risk assessment (SomaSignal) indicating high cardiovascular risk despite intermediate Framingham score. This prompts more aggressive preventive interventions including statin therapy and closer monitoring. Subsequent coronary calcium scoring confirms presence of early coronary disease.^54^
Case 5: Transplant Medicine
A 58-year-old kidney transplant recipient with stable creatinine undergoes protocol biopsy with transcriptome analysis revealing molecular signatures of subclinical rejection. Immunosuppression adjustment prevents clinical rejection and preserves long-term graft function.^55^
Practical Implementation Strategies for Physicians
Knowledge Development
- Identify Relevant Applications: Focus on omics applications most relevant to your specialty.^56^
- Utilize Available Resources: Organizations like CPIC, ClinGen, and specialty societies provide implementation guidance.^57^
- Continuing Education: Pursue targeted education in genomic medicine and other omics technologies through available CME programs.^58^
Clinical Workflow Integration
- Start Small: Begin with well-established applications (e.g., pharmacogenomics for common medications).^59^
- Leverage Clinical Decision Support: Implement point-of-care tools that integrate omics data into clinical workflows.^60^
- Establish Clear Pathways: Develop protocols for appropriate test ordering, interpretation, and clinical action.^61^
Team-Based Approaches
- Multidisciplinary Collaboration: Engage appropriate specialists (medical genetics, molecular pathology, bioinformatics).^62^
- Utilize Genetic Counselors: These professionals are invaluable for test selection, consent processes, and result interpretation.^63^
- Build Local Expertise: Develop "genomics champions" within practice groups or institutions.^64^
Future Directions
The clinical implementation of omics technologies continues to evolve rapidly. Key developments on the horizon include:
-
Point-of-Care Testing: Miniaturization and automation are bringing some omics technologies closer to the bedside, enabling real-time clinical decisions.^65^
-
Artificial Intelligence Integration: Machine learning approaches are improving the interpretation of complex multi-omics data, potentially leading to more accurate, clinically actionable insights.^66^
-
Dynamic Monitoring: Longitudinal omics profiling may enable detection of disease states before clinical symptoms emerge, shifting medicine toward a more preventive paradigm.^67^
-
Expanded Pharmacogenomics: Preemptive testing may become standard, with patients' genetic information integrated into electronic health records for lifelong medication guidance.^68^
-
Population Screening: As costs decrease and evidence accumulates, preventive genomic screening may become routine, identifying risk factors earlier and enabling targeted prevention.^69^
Conclusion
Omics technologies are transforming clinical practice across medical specialties by enabling more precise diagnosis, personalized treatment selection, and improved risk stratification. While barriers to implementation remain—including knowledge gaps, workflow integration challenges, and reimbursement issues—the clinical value of these approaches is increasingly well-established.
For practicing physicians, engaging with omics technologies does not require becoming a molecular biologist. Rather, it involves understanding the clinical applications relevant to one's specialty, developing the ability to appropriately select tests and interpret results, and establishing collaborative relationships with specialists in genetics, pathology, and bioinformatics.
As these technologies continue to mature and become more accessible, integrating omics-based approaches into clinical practice will become not just an option but a standard of care for providing optimal, personalized patient management.
References
-
Hasin Y, Seldin M, Lusis A. Multi-omics approaches to disease. Genome Biol. 2017;18(1):83.
-
Aronson SJ, Rehm HL. Building the foundation for genomics in precision medicine. Nature. 2015;526(7573):336-342.
-
Manolio TA, Abramowicz M, Al-Mulla F, et al. Global implementation of genomic medicine: We are not alone. Sci Transl Med. 2015;7(290):290ps13.
-
Green ED, Gunter C, Biesecker LG, et al. Strategic vision for improving human health at The Forefront of Genomics. Nature. 2020;586(7831):683-692.
-
Clark MM, Stark Z, Farnaes L, et al. Meta-analysis of the diagnostic and clinical utility of genome and exome sequencing and chromosomal microarray in children with suspected genetic diseases. NPJ Genom Med. 2018;3:16.
-
Splinter K, Adams DR, Bacino CA, et al. Effect of genetic diagnosis on patients with previously undiagnosed disease. N Engl J Med. 2018;379(22):2131-2139.
-
Relling MV, Klein TE. CPIC: Clinical Pharmacogenetics Implementation Consortium of the Pharmacogenomics Research Network. Clin Pharmacol Ther. 2011;89(3):464-467.
-
Stadler ZK, Maio A, Chakravarty D, et al. Therapeutic implications of germline testing in patients with advanced cancers. J Clin Oncol. 2021;39(24):2698-2709.
-
Mullard A. NCI-MATCH trial pushes cancer umbrella trial paradigm. Nat Rev Drug Discov. 2015;14(8):513-515.
-
Mangat PK, Halabi S, Bruinooge SS, et al. Rationale and Design of the Targeted Agent and Profiling Utilization Registry Study. JCO Precis Oncol. 2018;2:PO.18.00298.
-
Gregg AR, Aarabi M, Klugman S, et al. Screening for autosomal recessive and X-linked conditions during pregnancy and preconception: a practice resource of the American College of Medical Genetics and Genomics (ACMG). Genet Med. 2021;23(10):1793-1806.
-
Wilson MR, Sample HA, Zorn KC, et al. Clinical metagenomic sequencing for diagnosis of meningitis and encephalitis. N Engl J Med. 2019;380(24):2327-2340.
-
Byron SA, Van Keuren-Jensen KR, Engelthaler DM, Carpten JD, Craig DW. Translating RNA sequencing into clinical diagnostics: opportunities and challenges. Nat Rev Genet. 2016;17(5):257-271.
-
Cuzick J, Dowsett M, Pineda S, et al. Prognostic value of a combined estrogen receptor, progesterone receptor, Ki-67, and human epidermal growth factor receptor 2 immunohistochemical score and comparison with the Genomic Health recurrence score in early breast cancer. J Clin Oncol. 2011;29(32):4273-4278.
-
Sparano JA, Gray RJ, Makower DF, et al. Adjuvant chemotherapy guided by a 21-gene expression assay in breast cancer. N Engl J Med. 2018;379(2):111-121.
-
Sweeney TE, Perumal TM, Henao R, et al. A community approach to mortality prediction in sepsis via gene expression analysis. Nat Commun. 2018;9(1):694.
-
Halloran PF, Reeve J, Madill-Thomsen KS, et al. The Molecular Diagnosis of Rejection in Liver Transplant Biopsies: First Results of the INTERLIVER Study. Am J Transplant. 2017;17(9):2444-2454.
-
Wingo AP, Liu Y, Gerasimov ES, et al. Integrating human brain proteomes with genome-wide association data implicates new proteins in Alzheimer's disease pathogenesis. Nat Genet. 2021;53(2):143-146.
-
Tebani A, Afonso C, Marret S, Bekri S. Omics-based strategies in precision medicine: toward a paradigm shift in inborn errors of metabolism investigations. Int J Mol Sci. 2016;17(9):1555.
-
Curtis JR, van der Helm-van Mil AH, Knevel R, et al. Validation of a novel multibiomarker test to assess rheumatoid arthritis disease activity. Arthritis Care Res. 2012;64(12):1794-1803.
-
Williams SA, Kivimaki M, Langenberg C, et al. Plasma protein patterns as comprehensive indicators of health. Nat Med. 2019;25(12):1851-1857.
-
Ueland FR, Desimone CP, Seamon LG, et al. Effectiveness of a multivariate index assay in the preoperative assessment of ovarian tumors. Obstet Gynecol. 2011;117(6):1289-1297.
-
Petricoin EF, Liotta LA. Clinical proteomics: application at the bedside. Contrib Nephrol. 2008;160:11-18.
-
Nomura F. Proteome-based bacterial identification using matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS): A revolutionary shift in clinical diagnostic microbiology. Biochim Biophys Acta. 2015;1854(6):528-537.
-
Wishart DS. Emerging applications of metabolomics in drug discovery and precision medicine. Nat Rev Drug Discov. 2016;15(7):473-484.
-
Mak CM, Lee HC, Chan AY, Lam CW. Inborn errors of metabolism and expanded newborn screening: review and update. Crit Rev Clin Lab Sci. 2013;50(6):142-162.
-
Vander Heiden MG, DeBerardinis RJ. Understanding the intersections between metabolism and cancer biology. Cell. 2017;168(4):657-669.
-
Ahlqvist E, Storm P, Käräjämäki A, et al. Novel subgroups of adult-onset diabetes and their association with outcomes: a data-driven cluster analysis of six variables. Lancet Diabetes Endocrinol. 2018;6(5):361-369.
-
Kaddurah-Daouk R, Weinshilboum RM; Pharmacometabolomics Research Network. Pharmacometabolomics: implications for clinical pharmacology and systems pharmacology. Clin Pharmacol Ther. 2014;95(2):154-167.
-
Tang WHW, Bäckhed F, Landmesser U, Hazen SL. Intestinal microbiota in cardiovascular health and disease: JACC state-of-the-art review. J Am Coll Cardiol. 2019;73(16):2089-2105.
-
Karczewski KJ, Snyder MP. Integrative omics for health and disease. Nat Rev Genet. 2018;19(5):299-310.
-
Kuruvilla ME, Lee FE, Lee GB. Understanding asthma phenotypes, endotypes, and mechanisms of disease. Clin Rev Allergy Immunol. 2019;56(2):219-233.
-
Price ND, Magis AT, Earls JC, et al. A wellness study of 108 individuals using personal, dense, dynamic data clouds. Nat Biotechnol. 2017;35(8):747-756.
-
Huang S, Chaudhary K, Garmire LX. More is better: recent progress in multi-omics data integration methods. Front Genet. 2017;8:84.
-
van El CG, Cornel MC, Borry P, et al. Whole-genome sequencing in health care: recommendations of the European Society of Human Genetics. Eur J Hum Genet. 2013;21(6):580-584.
-
Burke W. Genetic tests: clinical validity and clinical utility. Curr Protoc Hum Genet. 2014;81:9.15.1-8.
-
Biesecker LG, Green RC. Diagnostic clinical genome and exome sequencing. N Engl J Med. 2014;370(25):2418-2425.
-
Stewart CM, Tsui DWY. Circulating tumor DNA for personalized cancer treatment. Clin Lab Med. 2018;38(2):293-310.
-
Clark MM, Hildreth A, Batalov S, et al. Diagnosis of genetic diseases in seriously ill children by rapid whole-genome sequencing and automated phenotyping and interpretation. Sci Transl Med. 2019;11(489):eaat6177.
-
Phillips KA, Deverka PA, Trosman JR, et al. Payer coverage policies for multigene tests. Nat Biotechnol. 2017;35(7):614-617.
-
Torkamani A, Andersen KG, Steinhubl SR, Topol EJ. High-definition medicine. Cell. 2017;170(5):828-843.
-
Knepper TC, Bell GC, Hicks JK, et al. Key lessons learned from Moffitt's Molecular Tumor Board: the Clinical Genomics Action Committee experience. Oncologist. 2017;22(2):144-151.
-
Richards S, Aziz N, Bale S, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405-424.
-
Kalia SS, Adelman K, Bale SJ, et al. Recommendations for reporting of secondary findings in clinical exome and genome sequencing, 2016 update (ACMG SF v2.0): a policy statement of the American College of Medical Genetics and Genomics. Genet Med. 2017;19(2):249-255.
-
Deignan JL, Chung WK, Kearney HM, et al. Points to consider in the reevaluation and reanalysis of genomic test results: a statement of the American College of Medical Genetics and Genomics (ACMG). Genet Med. 2019;21(6):1267-1270.
-
Appelbaum PS, Parens E, Waldman CR, et al. Models of consent to return of incidental findings in genomic research. Hastings Cent Rep. 2014;44(4):22-32.
-
Berkman BE, Wendler D. The interaction between bioethics and data science: the case of genomic privacy. J Law Med Ethics. 2022;50(2):318-326.
-
Popejoy AB, Fullerton SM. Genomics is failing on diversity. Nature. 2016;538(7624):161-164.
-
Wynn J, Martinez J, Duong J, et al. Research participants' preferences for hypothetical secondary results from genomic research. J Genet Couns. 2017;26(4):841-851.
-
Phillips KA, Trosman JR, Deverka PA, et al. Insurance coverage for genomic tests. Science. 2018;360(6386):278-279.
-
French CE, Delon I, Dolling H, et al. Whole genome sequencing reveals that genetic conditions are frequent in intensively ill children. Intensive Care Med. 2019;45(5):627-636.
-
Shaw AT, Solomon BJ, Besse B, et al. ALK resistance mutations and efficacy of lorlatinib in advanced anaplastic lymphoma kinase-positive non-small-cell lung cancer. J Clin Oncol. 2019;37(16):1370-1379.
-
Wilson MR, O'Donovan BD, Gelfand JM, et al. Chronic meningitis investigated via metagenomic next-generation sequencing. JAMA Neurol. 2018;75(8):947-955.
-
Ganz P, Heidecker B, Hveem K, et al. Development and validation of a protein-based risk score for cardiovascular outcomes among patients with stable coronary heart disease. JAMA. 2016;315(23):2532-2541.
-
Halloran PF, Famulski KS, Reeve J. Molecular assessment of disease states in kidney transplant biopsy samples. Nat Rev Nephrol. 2016;12(9):534-548.
-
Korf BR, Berry AB, Limson M, et al. Framework for development of physician competencies in genomic medicine: report of the competencies working group of the inter-society coordinating committee for physician education in genomics. Genet Med. 2014;16(11):804-809.
-
Knoppers BM, Zawati MH, Sénécal K. Return of genetic testing results in the era of whole-genome sequencing. Nat Rev Genet. 2015;16(9):553-559.
-
McGrath S, Ghersi D. Building towards precision medicine: empowering medical professionals for the next revolution. BMC Med Genomics. 2016;9(1):23.
-
Weitzel KW, Alexander M, Bernhardt BA, et al. The IGNITE network: a model for genomic medicine implementation and research. BMC Med Genomics. 2016;9:1.
-
Masys DR, Jarvik GP, Abernethy NF, et al. Technical desiderata for the integration of genomic data into electronic health records. J Biomed Inform. 2012;45(3):419-422.
-
Vassy JL, Christensen KD, Schonman EF, et al. The impact of whole-genome sequencing on the primary care and outcomes of healthy adult patients: A pilot randomized trial. Ann Intern Med. 2017;167(3):159-169.
-
van der Wouden CH, Carere DA, Maitland-van der Zee AH, et al. Consumer perceptions of interactions with primary care providers after direct-to-consumer personal genomic testing. Ann Intern Med. 2016;164(8):513-522.
-
Haga SB, Mills R, Pollak KI, et al. Developing patient-provider communication tools for cancer genomic testing. Commun Med. 2014;11(3):323-327.
-
Manolio TA, Chisholm RL, Ozenberger B, et al. Implementing genomic medicine in the clinic: the future is here. Genet Med. 2013;15(4):258-267.
-
Agarwal A, Antal CE, Shrivastava P, et al. Expanding molecular diagnostics to rare and emerging pathogens. Diagnostics (Basel). 2020;10(9):636.
-
Bumgarner R, Yeung KY. Methods for the inference of biological pathways and networks. Methods Mol Biol. 2009;541:225-245.
-
Karczewski KJ, Snyder MP. Integrative omics for health and disease. Nat Rev Genet. 2018;19(5):299-310.
-
Van Driest SL, Shi Y, Bowton EA, et al. Clinically actionable genotypes among 10,000 patients with preemptive pharmacogenomic testing. Clin Pharmacol Ther. 2014;95(4):423-431.
-
Schwartz MLB, McCormick CZ, Lazzeri AL, et al. A model for genome-first care: returning secondary genomic findings to participants and their healthcare providers in a large research cohort. Am J Hum Genet. 2018;103(3):328-337.