Harvey Cushing/John Hay Whitney Medical Library

  • Collections
  • Research Help

YSN Doctoral Programs: Steps in Conducting a Literature Review

  • Biomedical Databases
  • Global (Public Health) Databases
  • Soc. Sci., History, and Law Databases
  • Grey Literature
  • Trials Registers
  • Data and Statistics
  • Public Policy
  • Google Tips
  • Recommended Books
  • Steps in Conducting a Literature Review

What is a literature review?

A literature review is an integrated analysis -- not just a summary-- of scholarly writings and other relevant evidence related directly to your research question.  That is, it represents a synthesis of the evidence that provides background information on your topic and shows a association between the evidence and your research question.

A literature review may be a stand alone work or the introduction to a larger research paper, depending on the assignment.  Rely heavily on the guidelines your instructor has given you.

Why is it important?

A literature review is important because it:

  • Explains the background of research on a topic.
  • Demonstrates why a topic is significant to a subject area.
  • Discovers relationships between research studies/ideas.
  • Identifies major themes, concepts, and researchers on a topic.
  • Identifies critical gaps and points of disagreement.
  • Discusses further research questions that logically come out of the previous studies.

APA7 Style resources

Cover Art

APA Style Blog - for those harder to find answers

1. Choose a topic. Define your research question.

Your literature review should be guided by your central research question.  The literature represents background and research developments related to a specific research question, interpreted and analyzed by you in a synthesized way.

  • Make sure your research question is not too broad or too narrow.  Is it manageable?
  • Begin writing down terms that are related to your question. These will be useful for searches later.
  • If you have the opportunity, discuss your topic with your professor and your class mates.

2. Decide on the scope of your review

How many studies do you need to look at? How comprehensive should it be? How many years should it cover? 

  • This may depend on your assignment.  How many sources does the assignment require?

3. Select the databases you will use to conduct your searches.

Make a list of the databases you will search. 

Where to find databases:

  • use the tabs on this guide
  • Find other databases in the Nursing Information Resources web page
  • More on the Medical Library web page
  • ... and more on the Yale University Library web page

4. Conduct your searches to find the evidence. Keep track of your searches.

  • Use the key words in your question, as well as synonyms for those words, as terms in your search. Use the database tutorials for help.
  • Save the searches in the databases. This saves time when you want to redo, or modify, the searches. It is also helpful to use as a guide is the searches are not finding any useful results.
  • Review the abstracts of research studies carefully. This will save you time.
  • Use the bibliographies and references of research studies you find to locate others.
  • Check with your professor, or a subject expert in the field, if you are missing any key works in the field.
  • Ask your librarian for help at any time.
  • Use a citation manager, such as EndNote as the repository for your citations. See the EndNote tutorials for help.

Review the literature

Some questions to help you analyze the research:

  • What was the research question of the study you are reviewing? What were the authors trying to discover?
  • Was the research funded by a source that could influence the findings?
  • What were the research methodologies? Analyze its literature review, the samples and variables used, the results, and the conclusions.
  • Does the research seem to be complete? Could it have been conducted more soundly? What further questions does it raise?
  • If there are conflicting studies, why do you think that is?
  • How are the authors viewed in the field? Has this study been cited? If so, how has it been analyzed?

Tips: 

  • Review the abstracts carefully.  
  • Keep careful notes so that you may track your thought processes during the research process.
  • Create a matrix of the studies for easy analysis, and synthesis, across all of the studies.
  • << Previous: Recommended Books
  • Last Updated: Jun 20, 2024 9:08 AM
  • URL: https://guides.library.yale.edu/YSNDoctoral

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings
  • My Bibliography
  • Collections
  • Citation manager

Save citation to file

Email citation, add to collections.

  • Create a new collection
  • Add to an existing collection

Add to My Bibliography

Your saved search, create a file for external citation management software, your rss feed.

  • Search in PubMed
  • Search in NLM Catalog
  • Add to Search

How is genetic testing evaluated? A systematic review of the literature

Affiliations.

  • 1 Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy. [email protected].
  • 2 Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.
  • 3 Harvard TH Chan School of Public Health, Boston, MA, USA.
  • 4 Ministry of Health, Rome, Italy.
  • 5 Department of Health Sciences, Division of Medical Genetics, University of Genova, Galliera Hospital, Genova, Italy.
  • PMID: 29422659
  • PMCID: PMC5945588
  • DOI: 10.1038/s41431-018-0095-5

Given the rapid development of genetic tests, an assessment of their benefits, risks, and limitations is crucial for public health practice. We performed a systematic review aimed at identifying and comparing the existing evaluation frameworks for genetic tests. We searched PUBMED, SCOPUS, ISI Web of Knowledge, Google Scholar, Google, and gray literature sources for any documents describing such frameworks. We identified 29 evaluation frameworks published between 2000 and 2017, mostly based on the ACCE Framework (n = 13 models), or on the HTA process (n = 6), or both (n = 2). Others refer to the Wilson and Jungner screening criteria (n = 3) or to a mixture of different criteria (n = 5). Due to the widespread use of the ACCE Framework, the most frequently used evaluation criteria are analytic and clinical validity, clinical utility and ethical, legal and social implications. Less attention is given to the context of implementation. An economic dimension is always considered, but not in great detail. Consideration of delivery models, organizational aspects, and consumer viewpoint is often lacking. A deeper analysis of such context-related evaluation dimensions may strengthen a comprehensive evaluation of genetic tests and support the decision-making process.

PubMed Disclaimer

Conflict of interest statement

The authors declare that they have no conflict of interest.

PRISMA flow diagram of the…

PRISMA flow diagram of the review process

Similar articles

  • Beyond the black stump: rapid reviews of health research issues affecting regional, rural and remote Australia. Osborne SR, Alston LV, Bolton KA, Whelan J, Reeve E, Wong Shee A, Browne J, Walker T, Versace VL, Allender S, Nichols M, Backholer K, Goodwin N, Lewis S, Dalton H, Prael G, Curtin M, Brooks R, Verdon S, Crockett J, Hodgins G, Walsh S, Lyle DM, Thompson SC, Browne LJ, Knight S, Pit SW, Jones M, Gillam MH, Leach MJ, Gonzalez-Chica DA, Muyambi K, Eshetie T, Tran K, May E, Lieschke G, Parker V, Smith A, Hayes C, Dunlop AJ, Rajappa H, White R, Oakley P, Holliday S. Osborne SR, et al. Med J Aust. 2020 Dec;213 Suppl 11:S3-S32.e1. doi: 10.5694/mja2.50881. Med J Aust. 2020. PMID: 33314144
  • A proposal of a new evaluation framework towards implementation of genetic tests. Pitini E, D'Andrea E, De Vito C, Rosso A, Unim B, Marzuillo C, Federici A, Di Maria E, Villari P. Pitini E, et al. PLoS One. 2019 Aug 5;14(8):e0219755. doi: 10.1371/journal.pone.0219755. eCollection 2019. PLoS One. 2019. PMID: 31381569 Free PMC article.
  • How can the evaluation of genetic tests be enhanced? Lessons learned from the ACCE framework and evaluating genetic tests in the United Kingdom. Sanderson S, Zimmern R, Kroese M, Higgins J, Patch C, Emery J. Sanderson S, et al. Genet Med. 2005 Sep;7(7):495-500. doi: 10.1097/01.gim.0000179941.44494.73. Genet Med. 2005. PMID: 16170241
  • The clinical effectiveness and cost-effectiveness of testing for cytochrome P450 polymorphisms in patients with schizophrenia treated with antipsychotics: a systematic review and economic evaluation. Fleeman N, McLeod C, Bagust A, Beale S, Boland A, Dundar Y, Jorgensen A, Payne K, Pirmohamed M, Pushpakom S, Walley T, de Warren-Penny P, Dickson R. Fleeman N, et al. Health Technol Assess. 2010 Jan;14(3):1-157, iii. doi: 10.3310/hta14030. Health Technol Assess. 2010. PMID: 20031087 Review.
  • Avoiding and identifying errors in health technology assessment models: qualitative study and methodological review. Chilcott J, Tappenden P, Rawdin A, Johnson M, Kaltenthaler E, Paisley S, Papaioannou D, Shippam A. Chilcott J, et al. Health Technol Assess. 2010 May;14(25):iii-iv, ix-xii, 1-107. doi: 10.3310/hta14250. Health Technol Assess. 2010. PMID: 20501062 Review.
  • The GenoVA study: Equitable implementation of a pragmatic randomized trial of polygenic-risk scoring in primary care. Vassy JL, Brunette CA, Lebo MS, MacIsaac K, Yi T, Danowski ME, Alexander NVJ, Cardellino MP, Christensen KD, Gala M, Green RC, Harris E, Jones NE, Kerman BJ, Kraft P, Kulkarni P, Lewis ACF, Lubitz SA, Natarajan P, Antwi AA. Vassy JL, et al. Am J Hum Genet. 2023 Nov 2;110(11):1841-1852. doi: 10.1016/j.ajhg.2023.10.001. Am J Hum Genet. 2023. PMID: 37922883 Free PMC article. Clinical Trial.
  • [Genomics in Public Health Scientific evidence and prospects for integration in the prevention practice]. Traversi D, Calabrò GE, Francese C, Franchitti E, Pulliero A, Spatera P, Izzotti A, Ventura CD, Lai A, Bergna A, Galli M, Zehender G, Tamburro M, Lombardi A, Salzo A, DE Dona R, D'Amico A, Viccione V, Ripabelli G, Baccolini V, Migliara G, Pitini E, Marzuillo C, DE Vito C, Pastorino R, Villari P, Boccia S. Traversi D, et al. J Prev Med Hyg. 2023 Jan 13;63(3 Suppl 2):E1-E29. doi: 10.15167/2421-4248/jpmh2022.63.3s2. eCollection 2023 Mar. J Prev Med Hyg. 2023. PMID: 36818497 Free PMC article. Italian. No abstract available.
  • Disclosure of Genetic Risk Factors for Alzheimer's Disease to Cognitively Healthy Individuals-From Current Practice towards a Personalised Medicine Scenario. Galluzzi S, Pievani M, Zanetti O, Benussi L, The Italian-DIAfN Working Group, Frisoni GB, Di Maria E. Galluzzi S, et al. Biomedicines. 2022 Dec 8;10(12):3177. doi: 10.3390/biomedicines10123177. Biomedicines. 2022. PMID: 36551936 Free PMC article. Review.
  • Effective and Efficient Delivery of Genome-Based Testing-What Conditions Are Necessary for Health System Readiness? Husereau D, Steuten L, Muthu V, Thomas DM, Spinner DS, Ivany C, Mengel M, Sheffield B, Yip S, Jacobs P, Sullivan T. Husereau D, et al. Healthcare (Basel). 2022 Oct 19;10(10):2086. doi: 10.3390/healthcare10102086. Healthcare (Basel). 2022. PMID: 36292532 Free PMC article.
  • Barriers to genetic testing in clinical psychiatry and ways to overcome them: from clinicians' attitudes to sociocultural differences between patients across the globe. Pinzón-Espinosa J, van der Horst M, Zinkstok J, Austin J, Aalfs C, Batalla A, Sullivan P, Vorstman J, Luykx JJ. Pinzón-Espinosa J, et al. Transl Psychiatry. 2022 Oct 11;12(1):442. doi: 10.1038/s41398-022-02203-6. Transl Psychiatry. 2022. PMID: 36220808 Free PMC article. Review.
  • Khoury MJ, Coates RJ, Evans JP. Evidence-based classification of recommendations on use of genomic tests in clinical practice: dealing with insufficient evidence. Genet Med. 2010;12:680–3. doi: 10.1097/GIM.0b013e3181f9ad55. - DOI - PubMed
  • Marzuillo C, De Vito C, D’Andrea E, Rosso A, Villari P. Predictive genetic testing for complex diseases: a public health perspective. QJM. 2014;107:93–97. doi: 10.1093/qjmed/hct190. - DOI - PMC - PubMed
  • Khoury MJ, Bowen MS, Burke W, et al. Current priorities for public health practice in addressing the role of human genomics in improving population health. Am J Prev Med. 2011;40:486–93. doi: 10.1016/j.amepre.2010.12.009. - DOI - PMC - PubMed
  • Boccia S, Federici A, Colotto M, Villari P. Implementation of Italian guidelines on public health genomics in Italy: a challenging policy of the NHS. Epidemiol Prev. 2014;38:29–34. - PubMed
  • Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions, version 5.1.0. The Cochrane Collaboration, 2011.

Publication types

  • Search in MeSH

Related information

Linkout - more resources, full text sources.

  • Europe PubMed Central
  • Nature Publishing Group
  • PubMed Central

Other Literature Sources

  • scite Smart Citations
  • MedlinePlus Health Information

full text provider logo

  • Citation Manager

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Advertisement

Advertisement

Predictors of Genetic Testing Decisions: A Systematic Review and Critique of the Literature

  • Review Paper
  • Published: 11 April 2014
  • Volume 23 , pages 263–288, ( 2014 )

Cite this article

jeff is conducting a literature review on the research on genetics

  • Kate Sweeny 1 ,
  • Arezou Ghane 1 ,
  • Angela M. Legg 2 ,
  • Ho Phi Huynh 1 &
  • Sara E. Andrews 1  

1450 Accesses

34 Citations

7 Altmetric

Explore all metrics

Genetic testing is increasingly available in medical settings and direct-to-consumer. However, the large and growing literature on genetic testing decisions is rife with conflicting findings, inconsistent methodology, and uneven attention across test types and across predictors of genetic testing decisions. Existing reviews of the literature draw broad conclusions but sacrifice nuanced analysis that with a closer look reveals far more inconsistency than homogeny across studies. The goals of this paper are to provide a systematic review of the empirical work on predictors of genetic testing decisions, highlight areas of consistency and inconsistency, and suggest productive directions for future research. We included all studies that provided quantitative analysis of subjective (e.g., perceived risk, perceived benefits of testing) and/or objective (e.g., family history, sociodemographic variables) predictors of genetic testing interest, intentions, or uptake, which produced a sample of 115 studies. From this review, we conclude that self-reported and test-related (as opposed to disorder-related or objective) predictors are relatively consistent across studies but that theoretically-driven efforts to examine testing interest across test types are sorely needed.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Subscribe and save.

  • Get 10 units per month
  • Download Article/Chapter or eBook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime

Price includes VAT (Russian Federation)

Instant access to the full article PDF.

Rent this article via DeepDyve

Institutional subscriptions

Similar content being viewed by others

jeff is conducting a literature review on the research on genetics

A Rapid Systematic Review of Outcomes Studies in Genetic Counseling

“i would rather have it done by a doctor”—laypeople’s perceptions of direct-to-consumer genetic testing (dtc gt) and its ethical implications, public awareness of direct-to-consumer genetic tests: findings from the 2013 u.s. health information national trends survey, explore related subjects.

  • Medical Ethics

Ajzen, I. (2002). Perceived behavioral control, self-efficacy, locus of control, and the theory of planned behavior. Journal of Applied Social Psychology, 32 , 665–683.

Article   Google Scholar  

Ajzen, I., & Fishbein, M. (1977). Attitude-behavior relations: a theoretical analysis and review of empirical research. Psychological Bulletin, 84 , 888–918.

Aktan-Collan, K., Mecklin, J.-P., Jarvinen, H., Nyström-Lahti, M., Peltomäki, P., Söderling, I., et al. (2000). Predictive genetic testing for hereditary non-polyposis colorectal cancer: uptake and long-term satisfaction. International Journal of Cancer (Predictive Oncology), 89 , 44–50.

Article   CAS   Google Scholar  

Andrews, L., Meiser, B., Apicella, C., & Tucker, K. (2004). Psychological impact of genetic testing for breast cancer susceptibility in women of Ashkenazi Jewish background: a prospective study. Genetic Testing, 8 , 241–247.

Andrykowski, M. A., Munn, R. K., & Studts, J. L. (1996). Interest in learning of personal genetic risk for cancer: a general population survey. Preventive Medicine, 25 , 527–536.

Article   CAS   PubMed   Google Scholar  

Bates, M. D., Quinn Griffin, M. T., Killion, C. M., & Fitzpatrick, J. J. (2011). African-American males’ knowledge and attitudes toward genetic testing and willingness to participate in genetic testing: a pilot study. Journal of National Black Nurses Association, 22 , 1–7.

Google Scholar  

Becker, M. H. (Ed.) (1974). The health belief model and personal health behavior. Health Education Monographs, 2, 324-473.

Bernhardt, B. A., Geller, G., Stauss, M., Helzlsouer, K. J., Stefanek, M., Wilcox, P. M., et al. (1997). Toward a model informed consent process for BRCA1 testing: a qualitative assessment of women’s attitudes. Journal of Genetic Counseling, 6 , 207–222.

Article   PubMed   Google Scholar  

Biesecker, B. B., Ishibe, N., Hadley, D. W., Giambarresi, T. R., Kase, R. G., Lerman, C., et al. (2000). Psychosocial factors predicting BRCA1/BRCA2 testing decisions in members of hereditary breast and ovarian cancer families. American Journal of Medical Genetics, 93 , 257–263.

Binedell, J., Soldan, J. R., & Harper, P. S. (1998). Predictive testing for Huntington’s disease: I. Predictors of uptake in South Wales. Clinical Genetics, 54 , 477–488.

Bloch, M., Fahy, M., Fox, S., & Hayden, M. R. (1989). Predictive testing for Huntington’s disease: II. Demographic characteristics, life-style patterns, attitudes, and psychosocial assessments of the first fifty-one test candidates. American Journal of Medical Genetics, 32 , 217–224.

Bosompra, K., Flynn, B. S., Ashikaga, T., Rairikar, C. J., Worden, J. K., & Solomon, L. J. (2000). Likelihood of undergoing genetic testing for cancer risk: a population-based study. Preventive Medicine, 30 , 155–166.

Bosompra, K., Ashikaga, T., Flynn, B. S., Worden, J. K., & Solomon, L. J. (2001). Psychosocial factors associated with the public’s willingness to pay for genetic testing for cancer risk: a structural equations model. Health Education Research, 16 , 157–172.

Botoseneanu, A., Alexander, J. A., & Banaszak-Holl, J. (2011). To test or not to test? The role of attitudes, knowledge, and religious involvment among U.S. adults on intent-to-obtain adult genetic testing. Health Education & Behavior, 38 , 617–628.

Bottorff, S. L., Ratner, P. A., & Balneaves, L. G. (2002). Women’s Interest in genetic testing for breast cancer risk: the influence of sociodemographics and knowledge. Cancer Epidemiology, Biomarkers & Prevention, 11 , 89–95.

Braithwaite, D., Sutton, S., & Steggles, N. (2002). Intention to participate in predictive genetic testing for hereditary cancer: the role of attitude toward uncertainty. Psychology & Health, 17 , 761–772.

Bratt, O., Damber, J.-E., Emanuelsson, M., Kristoffersson, U., Lundgren, R., Olsson, H., et al. (2000). Risk perception, screening practice and interest in genetic testing among unaffected men in families with hereditary prostate cancer. European Journal of Cancer, 36 , 235–241.

Bunn, J. Y., Bosompra, K., Ashikaga, T., Flynn, B. S., & Worden, J. K. (2002). Factors influencing intention to obtain a genetic test for colon cancer risk: a population-based study. Preventive Medicine, 34 , 567–577.

Cameron, L. D., & Diefenbach, M. A. (2001). Responses to information about psychosocial consequences of genetic testing for breast cancer susceptibility: influences of cancer worry and risk perception. Journal of Health Psychology, 6 , 47–59.

Cameron, L. D., & Reeve, J. (2006). Risk perceptions, worry, and attitudes about genetic testing for breast cancer susceptibility. Psychology & Health, 21 , 211–230.

Cameron, L. D., Sherman, K. A., Marteau, T. M., & Brown, P. M. (2009). Impact of genetic risk information and type of disease on perceived risk, anticipated affect, and expected consequences of genetic tests. Health Psychology, 28 , 307–316.

Cappelli, M., Surh, L., Humphreys, L., Verma, S., Logan, D., Hunter, A., et al. (1999). Psychological and social determinants of women’s decisions to undergo genetic counseling and testing for breast cancer. Clinical Genetics, 55 , 419–430.

Cappelli, M., Surh, L., Humphreys, L., Verma, S., Logan, D., & Allanson, J. (2001). Measuring women’s preferences for breast cancer treatments and BRCA1/BRCA2 testing. Quality of Life Research, 10 , 595–607.

Cappelli, M., Hunter, A. G. W., Stern, H., Humphreys, L., Van Houten, L., O’Rourke, K., et al. (2002). Participation rates of Ashkenazi Jews in a colon cancer community-based screening/prevention study. Clinical Genetics, 61 , 104–114.

Chaliki, H., Loader, S., Levenkron, J. C., Logan-Young, W., Hall, W. J., & Rowley, P. T. (1995). Women’s receptivity to testing for a genetic susceptibility to breast cancer. American Journal of Public Health, 85 , 1133–1135.

Article   CAS   PubMed Central   PubMed   Google Scholar  

Cherkas, L. F., Harris, J. M., Levinson, E., Spector, T. D., & Prainsack, B. (2010). A survey of UK public interest in internet-based personal genome testing. PLoS One, 5 , e13473.

Article   PubMed Central   PubMed   CAS   Google Scholar  

Codori, A.-M., Hanson, R., & Brandt, J. (1994). Self-selection in predictive testing for Huntington’s disease. American Journal of Medical Genetics, 54 , 167–173.

Codori, A. M., Petersen, G. M., Miglioretti, D. L., Larkin, E. K., Bushey, M. T., Young, C., et al. (1999). Attitudes toward colon cancer gene testing: factors predicting test uptake. Cancer Epidemiology, Biomarkers & Prevention, 8 , 345–351.

CAS   Google Scholar  

Cragun, D., Malo, T. L., Pal, T., Shibata, D., & Vadaparampil, S. T. (2012). Colorectal cancer survivors’ interest in genetic testing for hereditary cancer: implications for universal tumor screening. Genetic Testing & Molecular Biomarkers, 16 , 493–499.

Craufurd, D., Kerzin-Storrar, L., Dodge, A., & Harris, R. (1989). Uptake of presymptomatic predictive testing for Huntington’s disease. The Lancet, 334 , 603–605.

Croyle, R. T., & Lerman, C. (1993). Interest in genetic testing for colon cancer susceptibility: cognitive and emotional correlates. Preventive Medicine, 22 , 284–292.

Croyle, R. T., Dutson, D. S., Tran, V. T., & Sun, Y.-C. (1995). Need for certainty and interest in genetic testing. Women’s Health: Research on Gender, Behavior, & Policy, 1 , 329–339.

Culler, D. D., Silberg, J., Vanner-Nicely, L., Ware, J. L., Jackson-Cook, C., & Bodurtha, J. (2002). Factors influencing men’s interest in gene testing for prostate cancer susceptibility. Journal of Genetic Counseling, 11 , 383–398.

Culver, J., Burke, W., Yasui, Y., Durfy, S., & Press, N. (2001). Participation in breast cancer genetic counseling: the influence of education level, ethnic background, and risk perception. Journal of Genetic Counseling, 10 , 215–231.

Cutler, S. J., & Hodgson, L. G. (2003). To test or not to test: Interest in genetic testing for Alzheimer’s disease among middle-aged adults. American Journal of Alzheimer’s Disease & Other Dementias, 18 , 9–20.

Cyr, A., Dunnagan, T. A., & Haynes, G. (2010). Efficacy of the health belief model for predicting intention to pursue genetic testing for colorectal cancer. Journal of Genetic Counseling, 19 , 174–186.

DiLorenzo, T. A., Schnur, J., Montgomery, G. H., Erblich, J., Winkel, G., & Bovbjerg, D. H. (2006). A model of disease-specific worry in heritable disease: the influence of family history, perceived risk and worry about other illnesses. Journal of Behavioral Medicine, 29 , 37–49.

Dugas, M. J., Gagnon, F., Ladouceur, R., & Freeston, M. H. (1998). Generalized anxiety disorder: a preliminary test of a conceptual model. Behaviour Research and Therapy, 36 , 215–226.

Durfy, S. J., Bowen, D. J., McTiernan, A., Sporleder, J., & Burke, W. (1999). Attitudes and interest in genetic testing for breast and ovarian cancer susceptibility in diverse groups of women in Western Washington. Cancer Epidemiology, Biomarkers & Prevention, 8 , 369–375.

Ellison, C. W., & Smith, J. (1991). Toward an integrative measure of health and well being. Journal of Psychological Theology, 19 , 35–48.

Etchegary, H. (2004). Psychological aspects of predictive genetic-test decisions: what do we know so far? Analyses of Social Issues & Public Policy, 4 , 13–31.

Etchegary, H., Capelli, M., Potter, B., Vloet, M., Graham, I., Walker, M., et al. (2010). Attitude and knowledge about genetics and genetic testing. Public Health & Genomics, 13 , 80–88.

Evers-Kiebooms, G., & Decruyenaere, M. (1998). Predictive testing for Huntington’s disease: a challenge for persons at risk and for professionals. Patient Education & Counseling, 35 , 15–26.

Evers-Kiebooms, G., Swerts, A., Cassiman, J. J., & Van Den Berghe, H. (1989). The motivation of at-risk individuals and their partners in deciding for or against predictive testing for Huntington’s disease. Clinical Genetics, 35 , 29–40.

Evers-Kiebooms, G., Welkenhuysen, M., Claes, E., Decruyenaere, M., & Denayer, L. (2000). The psychological complexity of predictive testing for late onset neurogenetic diseases and hereditary cancers: Implications for multidisciplinary counselling and for genetic education. Social Science & Medicine, 51 , 831-841.

Fisher, A., Bonner, C., Biankin, A., & Juraskova, I. (2012). Factors influencing intention to undergo whole genome screening in future healthcare: a single-blind parallel-group randomised trial. Preventive Medicine, 55 , 514–520.

Foster, C., Evans, D. G. R., Eeles, R., Eccles, D., Ashley, S., Brooks, L., et al. (2004). Non-uptake of predicitve testing for BRCA1/2 among relatives of known carriers: attributes, cancer, worry, and barriers to testing in a multicenter clinical cohort. Genetic Testing, 8 , 23–29.

Frost, S., Myers, L. B., & Newman, S. P. (2001). Genetic screening for Alzheimer’s disease: what factors predict intentions to take a test? Behavioral Medicine, 27 (3), 101–109.

Fulda, K. G., & Lykens, K. (2006). Ethical issues in predictive genetic testing: a public health perspectives. Journal of Medical Ethics, 32 , 143–147.

Glanz, K., Grove, J., Lerman, C., Gotay, C., & Le Marchand, L. (1999). Correlates of intentions to obtain genetic counseling and colorectal cancer gene testing among at-risk relatives from three ethnic groups. Cancer Epidemiology, Biomarkers & Prevention, 8 , 329–336.

Godard, B., Pratte, A., Dumont, M., Simard-Lebrun, A., & Simard, J. (2007). Factors associated with an individual’s decision to withdraw from genetic testing for breast and ovarian cancer susceptibility: implications for counseling. Genetic Testing, 11 , 45–54.

Gooding, H. C., Organista, K., Burack, J., & Biesecker, B. B. (2006). Genetic susceptibility testing from a stress and coping perspective. Social Science & Medicine, 62 , 1880–1890.

Graham, I. D., Logan, D. M., Hughes-Benzie, R., Evans, W. K., Perras, H., McAuley, L. M., et al. (1998). How interested is the public in genetic testing for colon cancer susceptibility? Report of a cross-sectional population survey. Cancer Prevention & Control, 2 , 167–172.

Gray, S. W., Hornik, R. C., Schwartz, J. S., & Armstrong, K. (2012). The impact of risk information exposure on women’s beliefs about direct-to-consumer genetic testing for BRCA mutations. Clinical Genetics, 81 , 29–37.

Gwyn, K., Vernon, S. W., & Conoley, P. M. (2003). Intention to pursue genetic testing for breast cancer among women due for screening mammography. Cancer Epidemiology, Biomarkers & Prevention, 12 , 96–102.

Hadley, D. W., Jenkins, J., Dimond, E., Nakahara, K., Grogan, L., Liewehr, D. J., et al. (2003). Genetic counseling and testing in families with hereditary nonpolyposis colorectal cancer. Archives of Internal Medicine, 163 , 573–582.

Hailey, B. J., Carter, C. L., & Burnett, D. R. (2000). Breast cancer attitudes, knowledge, and screening behavior in women with and without a family history of breast cancer. Health Care for Women International, 21 , 701–715.

Hall, J., Gray, S., A’Hern, R., Shanley, S., Watson, M., Kash, K., et al. (2009). Genetic testing for BRCA1: effects of a randomised study of knowledge provision on interest in testing and long term test uptake; implications for the NICE guidelines. Familial Cancer, 8 , 5–13.

Harel, A., Abuelo, D., & Kazura, A. (2003). Adolescents and genetic testing: what do they think about it? Journal of Adolescent Health, 33 , 489–494.

Helmes, A. W. (2002). Application of the protective motivation theory to genetic testing for breast cancer risk. Preventive Medicine, 35 , 453–462.

Helmes, A. W., Culver, J. O., & Bowen, D. J. (2006). Results of a randomized study of telephone versus in-person breast cancer risk counseling. Patient Education & Counseling, 64 , 96–103.

Hiraki, S., Chen, C. A., Roberts, J. S., Cupples, L. A., & Green, R. C. (2009). Perceptions of familial risk in those seeking genetic risk assessment for Alzheimer’s disease. Journal of Genetic Counseling, 18 , 130–136.

Article   PubMed Central   PubMed   Google Scholar  

Holloway, S. M., Bernhard, B., Campbell, H., & Lam, W. W. K. (2008). Uptake of testing for BRCA1/2 mutations in South East Scotland. European Journal of Human Genetics, 16 , 906–912.

Hughes, C., Gomez-Caminero, A., Benkendorf, J., Kerner, J., Isaacs, C., Barter, J., et al. (1997). Ethnic differences in knowledge and attitudes about BRCA1 testing in women at increased risk. Patient Education & Counseling, 32 , 51–62.

Jacobsen, P. B., Valdimarsdottir, H. B., Brown, K. L., & Offitt, K. (1997). Decision-making about genetic testing among women at familiar risk for breast cancer. Psychosomatic Medicine, 59 , 459–466.

Jacopini, G. A., D’Amico, R., & Vivona, G. (1992). Attitudes of persons at risk and their partners toward predictive testing. Birth Defects, 28 , 113–117.

CAS   PubMed   Google Scholar  

Janz, N., & Becker, M. (1984). The health belief model: a decade later. Health Education Quarterly, 11 , 1–47.

Julian-Reynier, C., Sobol, H., Sevilla, C., Nogues, C., & Bourret, P. (2000). Uptake of hereditary breast/ovarian cancer genetic testing in a French national sample of BRCA1 families. Psycho-Oncology, 9 , 504–510.

Kasparian, N. A., Meiser, B., Butow, P. N., Simpson, J. P., & Mann, G. J. (2009). Genetic testing for melanoma risk: a prospective cohort study of uptake and outcomes among Australian families. Genetic Medicine, 11 , 265–278.

Kelly, K., Leventhal, H., Andrykowski, M., Toppmeyer, D., Much, J., Dermody, J., et al. (2004). The decision to test in women receiving genetic counseling for BRCA1 and BRCA2 mutations. Journal of Genetic Counseling, 13 , 237–257.

Keogh, L. A., Southey, M. C., Maskiell, J., Young, M. A., Gaff, C. L., Kirk, J., et al. (2004). Uptake of offer to receive genetic information about BRCA1 and BRCA2 mutations in an Australian population-based study. Cancer Epidemiology, Biomarkers & Prevention, 13 , 2258–2263.

Kinney, A. Y., Choi, Y.-A., DeVellis, B., Kobetz, E., Millikan, R. C., & Sandler, R. S. (2000). Interest in genetic testing among first-degree relatives of colorectal cancer patients. American Journal of Preventive Medicine, 18 , 249–252.

Kinney, A. Y., Croyle, R. T., Dudley, W. N., Bailey, C. A., Pelias, M. K., & Neuhausen, S. L. (2001). Knowledge, attitudes, and interest in breast-ovarian cancer gene testing: a survey of a large African-american kindred with a BRCA1 mutation. Preventive Medicine, 33 , 543–551.

Klitzman, R., Thorne, D., Williamson, J., & Marder, K. (2007). The roles of family members, health care workers, and others in decision-making processes about genetic testing among individuals at risk for Huntington disease. Genetic Medicine, 9 , 358–371.

Laegsgaard, M. M., Kristensen, A. S., & Mors, O. (2009). Potential consumers’ attitudes toward psychiatric genetic research and testing and factors influencing their intentions to test. Genetic Testing & Molecular Biomarkers, 13 , 57–65.

Lee, S. C., Bernhardt, B. A., & Helzlsouer, K. J. (2002). Utilization of BRCA1/2 genetic testing in the clinical setting. Cancer, 94 , 1876–1885.

Lerman, C., Narod, S., Schulman, K., Hughes, C., Gomez-Caminero, A., Bonney, G., et al. (1996). BRCA1 Testing in families with hereditary breast-ovarian cancer: a prospective study of patient decision making and outcomes. JAMA, 275 , 1885–1892.

Lerman, C., Schwartz, M. D., Lin, T. H., Hughes, C., Narod, S., & Lynch, H. T. (1997). The influence of psychological distress on use of genetic testing for cancer risk. Journal of Consulting and Clinical Psychology, 65 , 414–420.

Lerman, C., Hughes, C., Trock, B. J., Myers, R. E., Main, D., Bonney, A., et al. (1999). Genetic testing in families with hereditary nonpolyposis colon cancer. JAMA, 281 , 1618–1622.

Lerman, C., Croyle, R. T., Tercyak, K. P., & Hamann, H. (2002). Genetic testing: psychological aspects and implications. Journal of Consulting and Clinical Psychology, 70 , 784–797.

Lipkus, I. M., Iden, D., Terrenoire, J., & Feaganes, J. R. (1999). Relationships among breast cancer concern, risk perceptions, and interest in genetic testing for breast cancer susceptibility among African-American women with and without a family history of breast cancer. Cancer Epidemiology, Biomarkers & Prevention, 8 , 533–539.

Lynch, H. T., Snyder, C. L., Lynch, J. F., Ghate, S., Narod, S. A., & Gong, G. (2009). Family information service participation increases the rates of mutation testing among members of families with BRCA1/2 mutations. The Breast Journal, 15 , 20–24.

Mastromauro, C., Myers, R. H., & Berkman, B. (1987). Attitudes toward presymptomatic testing in Huntington disease. American Journal of Medical Genetics, 26 , 271–282.

McGuire, A. L., Diaz, C. M., Wang, T., & Hilsenbeck, S. G. (2009). Social networkers’ attitudes toward direct-to-consumer personal genome testing. The American Journal of Bioethics, 9 , 3–10.

Meijers-Heijboer, E. J., Verhoog, L. C., Brekelmans, C. T. M., Seynaeve, C., Tilanus-Linthorst, M. M. A., Wagner, A., et al. (2000). Presymptomatic DNA testing and prophylactic surgery in families with a BRCA1 or BRCA2 mutation. The Lancet, 355 , 2015–2020.

Meiser, B. (2005). Psychological impact of genetic testing for cancer susceptibility: an update of the literature. Psycho-Oncology, 14 , 1060–1074.

Meiser, B., & Dunn, S. (2000). Psychological impact of genetic testing for Huntington’s disease: an update of the literature. Journal of Neurology, Neurosurgery & Psychiatry, 69 , 574–578.

Meiser, B., Butow, P., Barratt, A., Suthers, G., Smith, M., Colley, A., et al. (2000). Attitudes to genetic testing for breast cancer susceptibility in women at increased risk of developing hereditary breast cancer. Journal of Medical Genetics, 37 , 472–476.

Meissen, G. J., & Berchek, R. L. (1987). Intentions to use predictive testing by those at risk for Huntington’s disease: implications for prevention. American Journal of Community Psychology, 16 , 261–277.

Metcalfe, K. A., Fan, I., McLaughlin, J., Risch, H. A., Rosen, B., Murphy, J., et al. (2009). Uptake of clinical genetic testing for ovarian cancer in Ontario: a population-based study. Gynecologic Oncology, 112 , 68–72.

Miller, S. M. (1987). Monitoring and blunting: validation of a questionnaire to assess styles of information seeking under threat. Journal of Personality and Social Psychology, 52 , 345–353.

Myers, R. E., Hyslop, T., Jennings-Dozier, K., Wolf, T. A., Burgh, D. Y., Diehl, J. A., et al. (2000). Intention to be tested for prostate cancer risk among African-American men. Cancer Epidemiology, Biomarkers & Prevention, 9 , 1323–1328.

National Human Genome Research Institute. (2011, October 13). An overview of the Human Genome Project. Retrieved from http://www.genome.gov/12011238 . Accessed 25 March 2013

National Institutes of Health (2013, November 4). Genetic testing: How it is used for healthcare. Retrieved from http://report.nih.gov/NIHfactsheets/ViewFactSheet.aspx?csid=43 . Accessed 25 March 2013

Nisbett, R. E., & Wilson, T. D. (1977). Telling more than we can know: verbal reports on mental processes. Psychological Review, 84 , 231–259.

Nordin, K., Björk, J., & Berglund, G. (2004). Factors influencing intention to obtain a genetic test for a hereditary disease in an affected group and in the general public. Preventive Medicine, 39 , 1107–1114.

Olaya, W., Esquivel, P., Wong, J. H., Morgan, J. W., Freeberg, A., Roy-Chowdhury, S., et al. (2009). Disparities in BRCA testing: when insurance coverage is not a barrier. American Journal of Surgery, 198 , 562.

Oster, E., Dorsey, E. R., Bausch, J., Shinaman, A., Kayson, E., Oakes, D., et al. (2008). Fear of health insurance loss among individuals at risk for Huntington’s disease. American Journal of Medical Genetics Part A, 146A , 2070–2077.

Paglierani, L. M., Kalkwarf, H. J., Rosenthal, S. L., Huether, C. A., & Wenstrup, R. J. (2003). The impact of test outcome certainty on interest in genetic testing among college women. Journal of Genetic Counseling, 12 , 131–150.

Petersen, G. M., Larkin, E., Codori, A., Wang, C., Booker, S. V., Bacon, J., Giardiello, F. M., & Boyd, P. A. (1999). Attitudes toward colon cancer gene testing: Survey of relatives of colon cancer patients. Cancer Epidemiology, Biomarkers & Prevention, 8 , 337-344.

Rahman, B., Meiser, B., Sachdev, P., Barlow-Stewart, K., Otlowski, M., Zilliacus, E., et al. (2012). To know or not to know: an update of the literature on the psychological and behavioral impact of genetic testing for Alzheimer disease risk. Genetic Testing & Molecular Biomarkers, 16 , 1–8.

Ramirez, A. G., Aparicio-Ting, F. E., Miguel, S., de Majors, S., & Miller, A. R. (2006). Interest, awareness, and perceptions of genetic testing among Hispanic family members of breast cancer survivors. Ethnicity & Disease, 16 , 398–403.

Reitz, F., Barth, J., & Bengel, J. (2004). Predictive value of breast cancer cognitions and attitudes toward genetic testing on women’s interest in genetic testing for breast cancer risk. Psycho-Social Medicine, 1 , 1–11.

Roberts, J. S. (2000). Anticipating response to predictive genetic testing for Alzheimer’s disease: a survey of first-degree relatives. The Gerontologist, 40 , 43–52.

Roberts, J. S., Barber, M., Brown, T. M., Cupples, L. A., Farrer, L. A., LaRusse, S. A., et al. (2004). Who seeks genetic susceptibility testing for Alzheimer’s disease? Findings from a multisite, randomized clinical trial. Genetic Medicine, 6 , 197–203.

Rogers, R. W. (1983). Cognitive and psychological processes in fear appeals and attitude change: A revised theory of protection motivation. In J. T. Cacioppo & R. E. Petty (Eds.), Social psychophysiology (pp. 153–176). New York: Guilford Press.

Romero-Hidalgo, S., Urraca, N., Parra, D., Villa, A. R., Lisker, R., & Carnevale, A. (2009). Attitudes and anticipated reactions to genetic testing for cancer among patients in Mexico City. Genetic Testing & Molecular Biomarkers, 13 , 477–483.

Ruddy, K. J., Gelber, S., Shin, J., Garber, J. E., Rosenberg, R., Przypysny, M., et al. (2010). Genetic testing in young women with breast cancer: results from a Web-based survey. Annals of Oncology, 21 , 741–747.

Salkovskis, P. M., Dennis, R., & Wroe, A. L. (1999). An experimental study of influences on the perceived likelihood of seeking genetic testing: “Nondirectiveness” may be misleading. Journal of Psychosomatic Research, 47 , 439–447.

Salkovskis, P. M., Rimes, K. A., Bolton, J., & Wroe, A. L. (2010). An experimental investigation of factors involved in the decision to undertake genetic testing for schizophrenia. Journal of Mental Health, 19 , 202–210.

Sanderson, S. C., Humphries, S. E., Hubbart, C., Hughes, E., Jarvis, M. J., & Wardle, J. (2008). Psychological and behavioural impact of genetic testing smokers for lung cancer risk: a phase II exploratory trial. Journal of Health Psychology, 13 , 481–494.

Sanderson, S. C., O’Neill, S. C., Bastian, L. A., Bepler, G., & McBride, C. M. (2010). What can interest tell us about uptake of genetic testing? Intention and behavior amongst smokers related to patients with lung cancer. Public Health Genomics, 13 , 116–124.

Scheier, M. F., & Carver, C. S. (1985). Optimism, coping, and health: assessment and implications of generalized outcome expectancies. Health Psychology, 4 , 219–247.

Schwartz, M. D., Hughes, C., Roth, J., Main, D., Peshkin, B. N., Isaacs, C., et al. (2000). Spiritual faith and genetic testing decisions among high-risk breast probands. Cancer Epidemiology, Biomarkers & Prevention, 9 , 381–385.

Segal, M. E., Polansky, M., & Sankar, P. (2007). Predictors of uptake of obesity genetic testing among affected adults. Human Genetics, 120 , 641–652.

Shiloh, S., Petel, Y., Papa, M., & Goldman, B. (1998). Motivations, perceptions and interpersonal differences associated with interest in genetic testing for breast cancer susceptibility among women at high and average risk. Psychology & Health, 13 , 1071–1086.

Shiloh, S., Ben-Sinai, R., & Keinan, G. (1999). Controllability, predictability, and information-seeking style on interest in predictive genetic testing. Personality and Social Psychology Bulletin, 25 , 1187–1195.

Smith, K. R., & Croyle, R. T. (1995). Attitudes toward genetic testing for colon cancer risk. American Journal of Public Health, 85 , 1435–1438.

Smith, R. J., & Hone, S. (2003). Genetic screening for deafness. Pediatric Clinics of North America, 50 , 315–329.

Smith, A. W., Dougall, A. L., Posluszny, D. M., Somers, T. J., Rubinstein, W. S., & Baum, A. (2008). Psychological distress and quality of life associated with genetic testing for breast cancer risk. Psycho-Oncology, 17 , 767–773.

Struewing, J. P., Lerman, C., Kase, R. G., Giambarresi, T. R., & Tucker, M. A. (1995). Anticipated uptake and impact of genetic testing in hereditary breast and ovarian cancer families. Cancer Epidemiology, Biomarkers & Prevention, 4 , 169–173.

Susswein, L. R., Skrzynia, C., Lange, L. A., Booker, J. K., Graham, M. L., III, & Evans, J. P. (2008). Increased uptake of BRCA1/2 genetic testing among African American women with a recent diagnosis of breast cancer. Journal of Clinical Oncology, 26 , 32–36.

Sweeny, K., & Legg, A. M. (2011). Predictors of interest in direct-to-consumer genetic testing. Psychology & Health, 26 , 1259–1272.

Tambor, E. S., Rimer, B. K., & Strigo, T. S. (1997). Genetic testing for breast cancer susceptibility: awareness and interest among women in the general population. American Journal of Medical Genetics, 68 , 43–49.

Tibben, A., Frets, P. G., van de Kamp, J. J., Niermeijer, M. F., Vegter van der Vlis, M., Roos, R. A., et al. (1993). Presymptomatic DNA testing for Huntington disease: pretest attitudes and expectations of applicants and their partners in the Dutch program. American Journal of Medical Genetics, 48 , 10–16.

Trippitelli, C. L., Jamison, K. R., Folstein, M. F., Bartko, J. J., & DePaulo, J. R. (1998). Pilot study on patients’ and spouses’ attitudes toward potential genetic testing for bipolar disorder. American Journal of Psychiatry, 155 , 899–904.

Ulrich, C. M., Kristal, A. R., White, E., Hunt, J. R., Durfy, S. J., & Potter, J. D. (1998). Genetic testing for cancer risk: a population survey on attitudes and intention. Community Genetics, 1 , 213–222.

Van der Steenstraten, I. M., Tibben, A., Roos, R. A., Van de Kamp, J. J., & Niermeijer, M. F. (1994). Predictive testing for Huntington disease: nonparticipants compared with participants in the Dutch program. American Journal of Human Genetics, 55 (4), 618.

PubMed Central   PubMed   Google Scholar  

Vernon, S. W., Gritz, E. R., Peterson, S. K., Perz, C. A., Marani, S., Amos, C. I., et al. (1999). Intention to learn results of genetic testing for hereditary colon cancer. Cancer Epidemiology, Biomarkers & Prevention, 8 , 353–360.

Wade, C. H., Shiloh, S., Woolford, S. W., Roberts, J. S., Alford, S. H., Marteau, T. M., et al. (2012). Modeling decisions to undergo genetic testing for susceptibility to common health conditions: an ancillary study of the Multiplex Initiative. Psychology & Health, 27 , 430–444.

Wallston, K. A., Malcarne, V. L., Flores, L., Hansdottir, I., Smith, C. A., Stein, M. J., et al. (1999). Does God determine your health? The God locus of health control scale. Cognitive Therapy and Research, 23 , 131–142.

Warner, B. J., Curnow, L. J., Polglase, A. L., & Debinski, H. S. (2005). Factors influencing uptake of genetic testing for colorectal cancer risk in an Australian Jewish population. Journal of Genetic Counseling, 14 , 387–394.

Webster, D. M., & Kruglanski, A. W. (1994). Individual differences in need for cognitive closure. Journal of Personality and Social Psychology, 67 , 1049–1062.

Welkenhuysen, M., Evers-Kiebooms, G., & Van den Berghe, H. (1997). Attitudes toward predictive testing for Alzheimer’s disease in a student population. Psychiatric Genetics, 7 , 121–126.

Welkenhuysen, M., Evers-Kiebooms, G., Decruyenaere, M., Claes, E., & Denayer, L. (2001). A community based study on intentions regarding predictive testing for hereditary breast cancer. Journal of Medical Genetics, 38 , 540–547.

Westmaas, J. L., & Woicik, P. B. (2005). Dispositional motivations and genetic risk feedback. Addictive Behaviors, 30 , 1524–1534.

Wilde, A., Meiser, B., Mitchell, P. B., Hadzi-Pavlovic, D., & Schofield, P. R. (2010). Community interest in predictive genetic testing for susceptibility to major depressive disorder in a large national sample. Psychological Medicine, 41 , 1605–1613.

Wilson, S., Ryan, A. V., Greenfield, S. M., Clifford, S. C., Holder, R. L., Pattison, H. M., et al. (2008). Self-testing for cancer: a community survey. BMC Cancer, 8 , 102.

Withrow, K. A., Burton, S., Arnos, K. S., Kalfoglou, A., & Pandya, A. (2008). Consumer motivations for pursuing genetic testing and their preferences for the provision of genetic services for hearing loss. Journal of Genetic Counseling, 17 , 252-260.

Wolff, K., Nordin, K., Brun, W., Berglund, G., & Kvale, G. (2011). Affective and cognitive attitudes, uncertainty avoidance and intention to obtain genetic testing: an extension of the theory of planned behaviour. Psychology & Health, 26 , 1143–1155.

Wroe, A. L., & Salkovskis, P. M. (1999). Factors influencing anticipated decisions about genetic testing: experimental studies. British Journal of Health Psychology, 4 , 19–40.

Wroe, A. L., & Salkovskis, P. M. (2000). The effects of non-directive questioning on an anticipated decision whether to undergo predictive testing for heart disease: an experimental study. Behavioral Research & Therapy, 38 , 389–403.

Yaniv, I., Benador, D., & Sagi, M. (2004). On not wanting to know and not wanting to inform others: choices regarding predictive genetic testing. Risk Decision & Policy, 9 , 317–336.

Download references

Conflict of Interest

Kate Sweeny declares that she has no conflict of interest.

Arezou Ghane declares that she has no conflict of interest.

Angela M. Legg declares that she has no conflict of interest.

Ho Phi Huynh declares that he has no conflict of interest.

Sara E. Andrews declares that she has no conflict of interest.

Human and Animal Rights

No animal or human studies were carried out by the authors for this article.

Author information

Authors and affiliations.

Department of Psychology, University of California, 900 University Ave., Riverside, CA, 92521, USA

Kate Sweeny, Arezou Ghane, Ho Phi Huynh & Sara E. Andrews

Department of Psychology, Pace University, 861 Bedford Rd, Pleasantville, NY, 10570, USA

Angela M. Legg

You can also search for this author in PubMed   Google Scholar

Corresponding author

Correspondence to Kate Sweeny .

Rights and permissions

Reprints and permissions

About this article

Sweeny, K., Ghane, A., Legg, A.M. et al. Predictors of Genetic Testing Decisions: A Systematic Review and Critique of the Literature. J Genet Counsel 23 , 263–288 (2014). https://doi.org/10.1007/s10897-014-9712-9

Download citation

Received : 13 May 2013

Accepted : 11 March 2014

Published : 11 April 2014

Issue Date : June 2014

DOI : https://doi.org/10.1007/s10897-014-9712-9

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Genetic testing
  • Decision-making
  • Systematic review
  • Direct-to-consumer
  • Find a journal
  • Publish with us
  • Track your research

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • J Med Internet Res
  • v.23(9); 2021 Sep

Logo of jmir

Exploring the Use of Genomic and Routinely Collected Data: Narrative Literature Review and Interview Study

Helen daniels.

1 Population Data Science, Swansea University, Swansea, United Kingdom

Kerina Helen Jones

Sharon heys, david vincent ford, associated data.

Summary of findings.

Advancing the use of genomic data with routinely collected health data holds great promise for health care and research. Increasing the use of these data is a high priority to understand and address the causes of disease.

This study aims to provide an outline of the use of genomic data alongside routinely collected data in health research to date. As this field prepares to move forward, it is important to take stock of the current state of play in order to highlight new avenues for development, identify challenges, and ensure that adequate data governance models are in place for safe and socially acceptable progress.

We conducted a literature review to draw information from past studies that have used genomic and routinely collected data and conducted interviews with individuals who use these data for health research. We collected data on the following: the rationale of using genomic data in conjunction with routinely collected data, types of genomic and routinely collected data used, data sources, project approvals, governance and access models, and challenges encountered.

The main purpose of using genomic and routinely collected data was to conduct genome-wide and phenome-wide association studies. Routine data sources included electronic health records, disease and death registries, health insurance systems, and deprivation indices. The types of genomic data included polygenic risk scores, single nucleotide polymorphisms, and measures of genetic activity, and biobanks generally provided these data. Although the literature search showed that biobanks released data to researchers, the case studies revealed a growing tendency for use within a data safe haven. Challenges of working with these data revolved around data collection, data storage, technical, and data privacy issues.

Conclusions

Using genomic and routinely collected data holds great promise for progressing health research. Several challenges are involved, particularly in terms of privacy. Overcoming these barriers will ensure that the use of these data to progress health research can be exploited to its full potential.

Introduction

The progression of genomics in the last few decades has been remarkable. Since 2001, when the Human Genome Project mapped and sequenced virtually every gene in the human genome, genetic sequencing technology has advanced rapidly in both the public and private domains. Next-generation sequencing costs have plummeted by almost 100%, and research opportunities have grown exponentially as a result [ 1 ]. For example, a simple search in the medical database, PubMed, shows that research on genomics has more than quadrupled since 2000, from around 340,000 published articles on this topic growing to 1.5 million by 2020. This increase has translated into quicker diagnoses, better outcomes, and more effective health care for patients [ 2 - 4 ]. Great strides have been made in cancer research, for example, where patients are now being treated according to their own or the tumor’s genomic data [ 5 ].

Being able to use genomic data in conjunction with routinely collected data holds even greater potential to advance knowledge by including factors wider in scope. Precision medicine requires that novel correlations of genotype, phenotype, and the environment be identified to inform new methods for diagnosing, treating, and preventing disease in a way that is responsive to the individual [ 5 ]. Knowledge of gene-environment interactions can also contribute on a population level by informing health and public health services in areas such as service planning, population genetic testing, disease prevention programs, and policy development [ 6 ]. Routinely collected data, electronic health records (EHRs) in particular, already hold vast amounts of clinical and environmental information on large numbers of people and preclude the need for lengthy and expensive data collection. Adding these phenotypic data to knowledge about a person’s genome can elucidate new knowledge, such as that about gene-environment and gene-drug interactions, and can thus provide a richer understanding of health and disease [ 7 ].

Increasing the use of genomic data for health research is a high government priority to understand and address the causes of disease. The potential of integrating genomic and routine data sets has been recognized in the United Kingdom by the Welsh Government [ 8 ] via their Genomics for Precision Medicine Strategy and by Genomics England [ 9 ] with the inception of the 100,000 genome project in 2018, both of whom are investigating ways to link genomic data and EHRs. From a more international perspective, the former president of the United States, Barack Obama, launched the Precision Medicine Initiative to improve individualized care by combining genomic data and EHRs with diet and lifestyle information from US citizens [ 10 ]. The UK Chief Medical Officer, Dame Sally Davies’s “genomic dream ” of mainstreaming genomic medicine into National Health Service (NHS) standard care is becoming ever closer, which means that data from a person’s genome will likely be directly recorded into EHRs, making this type of research far more accessible [ 11 ].

As this field prepares to move forward, it is important to take stock of the current state of play in order to highlight new avenues for development, identify challenges, and ensure that adequate data governance models are in place for safe and socially acceptable progress. Previous work has examined the benefits and logistical challenges of integrating genomic and routinely collected data in health care practice, but less is known of this specifically in a research setting [ 6 , 12 , 13 ]. Therefore, our objective is to add to this literature by conducting a narrative literature review and a series of interviews that would provide an outline of the use of genomic data alongside routinely collected data in health research to date. It focuses on the types of data that have been used, the role of routinely collected data in these studies, the data sources, how researchers access the data, and the challenges surrounding their use. This will inform further work in developing a framework for working with genomic and routinely collected data [ 14 ].

Literature Review

First, we conducted a literature search of research that used genomic data in conjunction with routinely collected data. We define routinely collected data as data collected as a matter of course and not specifically for research [ 8 ]. Genomic data refer to the data generated after processing a person’s genome, in full or in part, for example, by sequencing [ 7 ]. Studies were eligible for inclusion if they had used both types of data in combination to answer a health research question. We included studies of any design published in either peer-reviewed journals or gray literature in the English language.

We searched the following databases to identify these studies: PubMed, Ovid, CINAHL, OpenGrey, CENTRAL, LILACS, and Web of Knowledge from inception until January 31, 2019. We also searched for books, gray literature, and websites. We used a piloted search strategy that included keywords representing genetic and routinely collected data, and the following is the search strategy used for PubMed and modified for use with the remaining databases:

  • (Gene OR Genetic* OR Genome* OR Genomic*)
  • ( Administration record* OR Anonymised OR Anonymized OR Anonymisation OR Anonymization OR Big data OR Clinical record* OR Data linkage OR Data mining OR Data science OR Education record* OR Ehealth OR EHR OR Electronic data OR Health record* OR Housing record* OR Encrypt* OR Insurance OR Linked data OR Medical record* OR Patient record* OR Prison record* OR Publically available OR Publicly available OR Register OR Registry OR Registries OR Routine data OR Routinely collected OR Safe haven )

This search in PubMed resulted in more than 50,000 hits. After initial pilot screenings, for example, by restricting to only publications within the last 10 years, it was clear that, given the number and heterogeneity of potentially relevant articles, neither a systematic review nor a meta-analysis would be practical. We took a pragmatic approach by scanning these articles to retrieve information on the following items until we reached data saturation, when no new information appeared in the text: types of genomic data; types and roles of routinely collected data; data sources; and data access models, that is, how researchers access the data. We chose examples from each criterion to ensure that we included a range of health conditions and presented them in a narrative format, which followed the format of the criteria given above.

To understand the use of genomic and routinely collected data in context, we recruited a purposive sample of individuals who have been involved in leading research projects using a combination of genomic and routinely collected data. We identified potential participants from the literature search outlined above and sent 19 interview invitations via email. Reminders were sent after 2 weeks, and if there was no response, we made no further contact. In total, 11 individuals agreed to participate in either an in-person or teleconferencing interview, depending on their geographical location. Participants were involved with the following projects: the Swansea Neurology Biobank (Swansea, Wales); Dementia Platform UK (Oxford, England); PsyCymru (Swansea, Wales); UK Biobank (Stockport, England); BC Generations Project (British Columbia, Canada); the Province of Ontario Neurodevelopmental Disorders (POND) Network, IC/ES (Ontario, Canada); Electronic and Medical Records and Genomics (eMERGE) Network (Vanderbilt, United States); and the Sax Institute’s 45 and Up study (New South Wales, Australia). We omitted any further information on the participants to maintain their anonymity. We developed interview questions with our advisory board: a group of UK geneticists and data scientists who were interested in using genomic data and our discussions centered around these:

  • What is the purpose of integrating the genetic data with health data?
  • What types of genetic data are being included?
  • Were there particular approvals you had to obtain? And if yes, what were these?
  • What were the main challenges encountered?
  • How did you address the challenges?
  • What is your main model for storing these data?
  • What access model(s) do you use? For example, safe room only, remote access, data released externally to researchers;
  • What are the conditions for access to data?

We followed up interviews by email if any answers needed clarification.

Textbox 1 provides a summary of the results of the literature review and interviews. Multimedia Appendix 1 Table S1 [ 15 - 32 ] provides a detailed summary of the results.

Summary of results.

Type of genomic data

  • Single nucleotide polymorphisms
  • Polygenic risk scores
  • Gene activity scores
  • DNA methylation status

Purpose of combining data

  • Genome-wide association studies
  • Phenome-wide association studies
  • Longitudinal studies
  • Candidate gene studies
  • Gene profiling studies
  • Exploratory studies

Types of routinely collected data

  • Electronic health records
  • Disease registry data
  • Disease registries
  • Mortality registers
  • Deprivation indices
  • Health insurance

Role of routinely collected data

  • Identifying cases and controls
  • Baseline data
  • Deep phenotyping
  • Long-term follow-up
  • Sociodemographic information

Sources of data

Governance models for data access

  • Publicly available on the web
  • Released to researchers
  • Data safe havens
  • Data collection
  • Data storage and costs
  • Technical and/or software issues
  • Data privacy and data protection laws

We included 19 studies in this literature review that provided broad examples of the different types of genomic and routinely collected data that can be used together to answer a health-related research question. The countries where this research was based were the United Kingdom [ 15 - 20 ], China [ 21 ], United States [ 22 - 29 ], Canada [ 19 , 30 , 31 ], and Australia [ 32 ].

Types of Genomic Data

Examples found of the types of genomic data used in these studies included single nucleotide polymorphisms (SNPs), gene activity scores, and DNA methylation status. The most frequently used were SNPs [ 16 , 17 , 19 , 23 - 29 ], which represent a single base-pair change in the DNA sequence and are highly granular; hence, they are popular in health research [ 33 ]. Research studies tend to refer to SNPs by their reference SNP number, a unique identifier given to a SNP, or a cluster of SNPs by the National Center for Biotechnology (NCBI) [ 34 ].

Polygenic risk scores are closely related to SNPs and were used in 2 of the studies included in this review [ 15 , 16 ]. These predict a person’s likelihood of developing a particular disease based on the cumulative effect of a number of genetic variants [ 15 ]. Our literature search found other instances of quantitative measures of gene activity combined with routine data, including the 21-gene Recurrence Score, which measures the activity of 21 genes (16 cancer-related and 5 reference) for patients with breast cancer [ 33 ] and the enzyme activity score for the CYP2D6 (cytochrome P450 family 2 subfamily D member 6) gene, which codes for an important drug-metabolizing enzyme, and is highly polymorphic in humans [ 34 ]. Rusiecki et al [ 29 ] measured changes in DNA methylation status in their participants to investigate whether this predicted posttraumatic stress disorder in US military service members.

Purpose of Combining Genomic and Routine Data

Conducting genome-wide association studies (GWAS) [ 15 , 17 - 19 , 21 ], phenome-wide association studies (PheWAS) [ 24 , 25 ], and a combination of both [ 26 , 27 ] were the main purposes of combining genomic and routine data. Both these methods use powerful statistical techniques to find associations between genetic variants (SNPs) and phenotypes, which can then be used to predict the genetic risk factors of disease, levels of gene expression, and even social and behavioral characteristics such as educational attainment, impulsivity, and recreational drug experimentation [ 35 , 36 ]. Given the number of tested associations, these studies require large sample sizes to yield enough statistical power to detect differences in genetic variation between cases and controls [ 37 ]. Other study designs included longitudinal studies [ 16 , 18 , 30 - 32 ], a candidate gene study [ 23 ], case control studies [ 19 , 29 ], a gene profiling study [ 22 ], and an exploratory study [ 28 ]. Each of these studies was designed either to identify genetic risk factors of the disease or to investigate drug safety or efficacy ( Multimedia Appendix 1 Table S1).

Types and Role of Routinely Collected Data

EHRs appear to be, by far, the most common form of routinely collected data used in the studies identified in this review. For the type of research discussed here, data in EHRs have been used to identify eligible participants, for phenotyping, and to provide long-term follow-up on specific health outcomes. These data are collected as a matter of course in health care systems, and depending on their country of origin, EHR content can vary, although usually this digital record will include the patient’s name, address, demographics, medical history, care preferences, lifestyle information (such as diet, exercise, and smoking status), and free-text notes [ 38 ]. An example of EHR data used in the identified studies was the International Classification of Disease (ICD) coding. This allows clinicians to record the status of a patient in a standardized way, whether it is for disease, disorder, injury, infection, or symptoms [ 39 ]. Hebbring et al [ 26 ] used ICD codes in EHRs to identify appropriate cases and controls for their PheWAS study of the HLA-DRB1*150 gene involved in immune regulation. Rusiecki et al [ 29 ] used ICD codes to identify cases with a postdeployment diagnosis of posttraumatic stress disorder in their study on the link between gene expression and posttraumatic stress disorder in US military service members.

EHRs were also used to describe and validate a phenotype of interest, which is particularly important for PheWAS studies. The large number of phenotypes used in these analyses need to be clearly defined to ensure that any associations made with genetic variants are precise and replicable [ 40 ]. For example, Breitenstein et al [ 23 ] used EHRs to define the type 2 diabetes phenotype needed for their candidate gene study based on diagnoses, medications, and laboratory tests. The algorithm used to achieve this was developed by the eMERGE Network (see below for more details on this organization) and has been used successfully many times since [ 41 ].

In addition to providing a baseline snapshot of the patient, EHRs are also longitudinal in nature, and this makes them ideally placed to provide long-term follow-up to study participants. An example of this is the Genetics and Psychosis (GAP) study, which looked at a purported association between a variant of the ZNF804A gene and poor outcomes after first-episode psychosis [ 20 ]. Using individual-level linkage between EHRs and genotype data, this study followed the clinical outcomes for 291 patients over a period of 2 years, and subsequently found strong evidence for their hypothesis.

Other examples of routine data used in genomic research include disease registry morbidity and mortality records [ 20 , 22 ]. Disease registries collect information on clinical outcomes and care for a specific patient population over time. EHRs often feed data into these registries, but registry data can also include patient-reported outcomes and other biometric data, and therefore provide a more holistic view of the patient than an EHR would in isolation, for example, the UK MS Register [ 42 ]. Routine data sets need not be individual-level or person-based to prove useful in genetic research. A case in point is the Scottish Index of Multiple Deprivation, which gives an indication of a geographical area’s socioeconomic deprivation based on data about employment, income, health, education, housing, crime, and access to services [ 43 ]. Clarke et al [ 15 ] were able to link participants’ postcodes to the Scottish Index of Multiple Deprivation and generate socioeconomic deprivation variables to investigate their association with polygenic risk scores for alcohol dependence.

Sources of Genomic and Routinely Collected Data

Although individual research projects often collect biological samples and generate their own genomic data for combination with routinely collected data [ 19 , 29 ], studies can also make use of the many sources of genomic and routinely collected data already available. Our interview participants spoke to us about the different data sources they used in their research and provided details on the use of these data and the participants of their projects. From this information, there seem to be two general categories of these sources: databanks and biobanks, the former where only the data are stored, and the latter, which store both biological samples and genomic data. Below are illustrative examples; therefore, this is not an exhaustive list.

Dementias Platform UK

Dementias Platform UK (DPUK) [ 16 ] is a data portal funded by the Medical Research Council that hosts the data of 2 million people from over 40 cohorts relevant to dementia research. Combining these data enhances the individual research power of each study and brings together knowledge from a number of stakeholders to facilitate and accelerate new discoveries. Examples of these cohorts are the GENetic Frontotemporal Dementia Initiative (GENFI; genotype data for GRN , MAPT, and C9ORF72 genes) and the Genetic and Environmental Risk in Alzheimer’s Disease (GERAD) Consortium (whole exome sequences) [ 16 ]. Presently, DPUK provides EHR linkage for Welsh participants via the Secure Anonymized Information Linkage (SAIL) databank [ 44 ].

UK Biobank [ 45 ] is a national resource with data from 500,000 participants aged between 40 and 69 years who have donated blood, urine, and saliva samples, have undergone a number of baseline measures, and have provided detailed health and behavioral information about themselves. Genomic data are available for 488,000 participants and comprise SNPs, genotypes, and haplotypes. The UK Biobank holds a number of routine data sets, including hospital inpatient episodes, cancer registrations, and deaths. Studies using the UK Biobank genomic and routinely collected data include genome-wide meta-analyses of depression [ 17 ] and identifying candidate gene and disease associations that could help predict adverse drug reactions [ 26 ].

Personal Genome Project

The Personal Genomes Project (PGP) is a databank founded in 2005 at Harvard University and now extends worldwide. It provides a web-based platform for individuals (over 100,000 people to date) to share their genomic data publicly, along with their EHRs, and other trait information to progress science without many of the governance restrictions of traditional research. Most of the genomic data on the PGP database are in SNP format, although files of raw sequence data are available for some participants [ 43 ].

eMERGE Network

The eMERGE Network is a consortium of American medical institutions whose goal is to use EHR data in combination with a variety of genomic data types to advance translational research. The network also releases its genomic data, including GWAS, whole genome, and whole exome sequence data, along with a subset of phenotypic elements to the broader community of researchers via the dbGaP—an NCBI [ 46 ] database of genotypes and phenotypes. Through this mechanism, any research project can be applied to the uploaded data. A wealth of publications have resulted from eMERGE’s work; these are available to view on the web [ 24 , 25 , 47 ].

POND Network

The POND network is an IC/ES [ 31 ] initiative based in Ontario, Canada, and involves a cohort of children and young people (the total number of the sample is approximately 3000) with a neurological development disorder, with a particular focus on autism. This is a highly phenotyped cohort with all participants having undergone multiple clinical tests, including those for attention-deficit/hyperactivity disorder, obsessive-compulsive disorder, and family history and demographic data. A subset (n=667) provided consent for linkage with administrative data held in IC/ES via health card numbers. The aim is to identify subgroups of autism based on the co-occurrence of other developmental conditions, other comorbidities, and health service use, and to characterize these groups based on clinical attributes and genomics. The network data are project-level only, although it is anticipated that linkage of genomic data to administrative and health data will become more routine in the future.

BC Generations Project

The BC Generations Project [ 48 ] is British Columbia’s (BC) largest ever health study and is part of a national initiative—the Canadian Partnership for Tomorrow Project—to aid researchers in answering questions about how environment, lifestyle, and genes contribute to cancer and other chronic diseases. Almost 30,000 participants were involved in the project, and they provided baseline information about their health, diet, lifestyle, and medical and family history. Many have also donated blood and urine samples, and the type of genomic data generated from these samples is based on the needs of the researchers (subject to approval). The BC cohort is one of several provincial cohorts that can be combined for national studies or can be linked to provincial administrative data via PopDataBC [ 49 ].

Sax Institute and the 45 and Up Study

Based in Australia, the main business of the Sax Institute is to manage the 45 and up study, which has been following 260,000 people for over 12 years who provide both routinely collected and self-reported health data [ 32 ]. The aim of this study was to collect samples from 50,000 individuals for full genome sequencing. The Sax Institute has a partnership with the Garvan Institute of Medical Research [ 50 ], which acts as a genome sequencing facility and makes data available for research subject to approval. The Garvan Institute retains all the genomic data, but with data linkage to the Sax Institute.

Data Governance Access Models

We surveyed the data access models and information governance systems of genomic and routine data sources identified by our literature search and interviews. From the additional information given to us by our interview participants, we were able to categorize data access models as follows: (1) publicly available on the web, (2) released to researchers, or accessed via a (3) data safe haven.

Publicly Available on the Web

There are a wealth of free, genomic data sources on the web, made available because of individual projects or from the pooled results of a variety of different projects. These data are either downloadable or viewable on the web. For this type of resource, data tend to be at the gene or variant level (eg, GIANT Consortium [ 51 ]; GWAS Catalog [ 52 ]), but some do hold individual-level data, eg, Database of Genomic Variants [ 53 ]. The Personal Genome Project, described above, is the only data source that provides identifiable, individual-level genomic data [ 43 ]. All other data were deidentified.

Released to Researchers

Currently, the most common way for genomic data to be accessed for research is through secure electronic file transfer to the researcher. This generally occurs after successful application to an internal review board (IRB) and signed data use agreement (DUA). All of the published research studies identified by our literature search used this model, unless the genomic data were generated specifically for that particular project. For instance, Cronin et al [ 24 ] received genomic data regarding 54 SNPs, and also demographic, vital sign, and billing data derived linked EHRs by the eMERGE network. Other biobanks using this model include Mayo Genome Consortia [ 23 ], China Kadoorie Biobank [ 21 ], UK Biobank [ 17 , 26 ], BioVu [ 24 ], and Generation Scotland [ 17 ], although access to BioVu data is only granted to Vanderbilt faculty members [ 54 ]. In addition to IRB and DUA procedures, the use of individual-level EHR data supplied by Generation Scotland also requires an application to the NHS Research Ethics Committee [ 55 ].

Data Safe Havens

The use of large-scale population data in health research has become increasingly popular in the last few decades, and this has seen the evolution of data safe havens as a way to ensure its safe and secure use. Lea et al [ 56 ] defined data safe havens as a system that invokes procedural, technical, and physical controls, including access to data within a secure environment (rather than data release), in order to safeguard the identities of people providing the data. Despite there being a greater tendency in the literature for releasing data to researchers, we have seen from our work with interview participants, a trend toward using a data safe haven system for both younger and more well-established organizations.

The PsyCymru study [ 18 ] and the Swansea Neurology Biobank [ 19 ] have deposited polygenic risk scores and SNPs, respectively, into the SAIL Databank [ 44 ], a data safe haven based at Swansea University. The SAIL Databank provides remote access to many linkable anonymized data sets, and both of these studies have used SAIL to link their genetic data and other phenotypic data to EHRs and other routinely collected data. These data are available only for project access, and currently cannot be shared. However, their intention is to make linked genetic and routinely collected data available for research in the near future [ 57 , 58 ].

The Sax Institute operates in a similar way to the SAIL Databank in that it provides access to genomic and health data via a virtual lab by remote access anywhere in the world. Data access requires approval by two data access committees: one at the Garvan Institute and one at the Sax Institute. The BC Generations Project said, “should the researcher access the project’s data via PopDataBC then they would only be allowed to use the data on within the secured research environment” (Participant 5, BC Generations Project).

For the UK Biobank, the current modus operandi releases anonymized data externally to researchers. However, they stated that this is unlikely to be sustainable because genomic data files are too large. The UK Biobank will likely be changing to a remote access model in the near future. The eMERGE Network also confirmed that they are experimenting with remote access into a data safe haven. One of our participants who used IC/ES data explained that “as the data are considered highly sensitive, access is only by an ICES analyst, with results provided to the project lead” (Participant 4, IC/ES).

Combining genomic and routine data does not come without its challenges. Our participants, who are currently working with these data, spoke to us about these different challenges during their interviews, and their experiences are summarized below.

Data Collection

A challenge described by several participants concerned data collection. Conducting long-term follow-up over long periods and at regular intervals means that participants need to be invited and reconsented to provide more blood and other health information. In addition, poor quality sequence alignment could render the samples useless, and with repeated use by researchers, blood samples will eventually become exhausted. Each of these issues necessitates a lengthy and expensive process of resampling thousands of individuals and imposes a burden on participants. A possible solution to this, as one participant suggested, is to “join up with others biobanks in order to work towards epidemiologically valid sample sizes” (Participant 1, 45 and Up Study)

Data Storage and Costs

Genomic data are huge, approximately 90 GB for the raw data of 1 whole genome, and this is often the source of technical issues surrounding its use [ 59 ]. One participant told us that their main challenge was data storage capacity and that “it may no longer be cost or space efficient for organisations to hold multiple datasets” (Participant 2, UK Biobank). They go on to explain that there is certainly a case to be made for storing genomic data as VCF files only, which keep record only of gene sequence variations and are much smaller and easier to work with. However, this restricts the type of analyses possible, particularly in the advent of new discoveries about the anatomy of the genome. There may be a need to find solutions regarding storage space for raw genomic data and for specialist platforms required to conduct analyses.

Technical and/or Software Issues

Many of the challenges faced by participants working with these data are technical in nature. Participant 3 (eMERGE) described creating a sequencing platform from the ground up, which was much longer than initially projected. Furthermore, they described that multiple sequencing centers needed harmonization, as well as the needs across sites and projects for network-wide data collection. Another participant found difficulties surrounding analysis software and analyst capacity, since this is a very specialized skill (Participant 4, IC/ES). Researchers often conduct genetic data analysis using publicly available, downloadable software applications, which are subject to frequent updates for improvements. This is a challenge to incorporate this software and keep it up to date. Several other participants spoke of similar software issues. Potential solutions discussed during our interviews were to upscale and have dedicated servers for genomic data, as well as to install specialist toolsets within the secure research environment.

Data Privacy and Data Protection Laws

Given the possibility of identifying individuals from genomic data [ 60 ], privacy is of primary concern. We were told that, for one research project using IC/ES data (Participant 4), the privacy approval group was concerned about identifiability due to the genomic data being unique, particularly where there are rare variants. This means that the genomic data contained in variant call files have been brought into the databank to show the feasibility of transfer but has to be integrated into the analytical platform.

The General Data Protection Regulation [ 61 ] in the United Kingdom states that if researchers are to rely on the lawful basis of consent to process medical genomic data, the reasons for doing so must be described in an explicit and transparent way. The rapid decrease in the cost of whole genome sequencing in the last few decades has opened many new avenues for genetic research, but this means that it is impossible to predict what this research will actually look like in the future [ 55 ]. One participant felt that this means they would need to seek consent from the participant for each new research proposal [ 62 ], rather than just once when their sample was taken. They expressed concerns that “continued and lengthy re-contact with participants was not only costly and difficult, it may also be invasive and burdensome” (Participant 11, Swansea Neurology Biobank). Also mentioned was Canada’s antidiscrimination laws (Bill S-201: Genetic Nondiscrimination Act) to avoid prejudice on the grounds of genetics (including insurance and employability) [ 63 ] (Participant 5, BC Generations Project). Several participants believed that the public acceptability of their work was important, but they “weren’t sure how to go about ensuring it” (Participant 7, Dementia Platform UK).

Principal Findings

Using genomic and routinely collected data holds great potential for health research. The genomic data we identified in our literature review included SNPs, polygenic risk scores, and gene activity scores. Routine data primarily consisted of EHRs, but we did find other routine data types including registry data and deprivation indices that had been combined with genomic data. This paper shows how genomic research has progressed in recent years—from basic GWA and PheWA studies—to more complex methodologies in which health records are linked to genomic data at the individual level. Associations between genetic variants and phenotypes, identification of drug targets, knowledge about drug toxicity, and effectiveness can all be studied from the combination of genomic and routine data and leveraged for public benefit.

The EHRs created during routinely collected care provide the large sample sizes needed for GWAS analysis without the need for costly and time-consuming prospective data collection. These larger samples allow for greater validity, generalizability, and yield adequate statistical power, while also minimizing participant burden and reducing attrition during follow-up [ 64 ]. In addition, these data sets are not limited to a circumscribed number of phenotypes in the way that a traditional research study might be [ 40 ]. This richness and diversity of EHR data means that the large number of phenotypes used in PheWAS can be very clearly defined (referred to as deep phenotyping), which enhances the accuracy of phenotype-genotype associations [ 40 ].

Most importantly, with regard to EHRs, perhaps, is the additional information that routinely collected data provide about an individual’s environment. This can include lifestyle factors, education, work history, pollution, and even traumatic events. Genetic determinism has long been rejected, and we now widely accept the powerful influence that lifestyle and the environment have on the way that our genes are expressed [ 65 - 67 ]. Precision medicine requires novel correlations not only between genotype and phenotype to be made but also with an individual’s environment to inform new methods for diagnosing, treating, and preventing disease in a way that is tailored to that individual. Linking genomic data to routine data promises to elucidate important findings for precision medicine research, which will enable researchers to understand the relationship between an individual’s genome and their complete life course [ 68 ]. More of these types of studies are needed for precision medicine to reach its full potential, to make the intricate genotype-phenotype associations needed to advance the understanding and treatment of disease.

From our interviews, we identified 3 main ways that researchers can access genomic and routine data: publicly available on the web, released to researchers, and via a data safe haven. We also observed that biobanks and databanks seem to be moving toward renouncing a data release model and instead favoring a data safe haven approach. Aside from potentially solving data storage issues, this will also help assuage privacy and governance concerns. Reidentification and disclosure from genomic data is possible [ 69 , 70 ] and can lead to many undesirable consequences: discrimination by health or life insurance companies, societal stigma, and the discovery of a genetic predisposition to a condition when one does not want to be told. This is complicated further because of the familial nature of genes, and disclosure could cause some devastating effects for biological relatives as well [ 71 - 73 ].

Despite this, we must keep in mind that simply because genomic data are unique, this in itself does not render it identifiable. Rather, the reidentification risk of genomic data depends on the way they are accessed, the type of analyses that are conducted, and the format in which the results are finally published [ 70 ]. This means that decisions to use certain data access models based on practicality and decreased costs are not sufficient. Genomic research relies heavily on human participation, and the public should be consulted to inform the way in which their data are accessed [ 74 ]. There is a plethora of activity taking place to consult the public about health research in general, and the success and acceptability of large-scale data research is owed, in part, to the extensive public engagement activities that have been taking place [ 75 , 76 ]. Organizations such as the Global Alliance for Genomics and Health [ 77 ] are engaging with the public on the use of genomic data in research, but there is still more work to be done regarding the use of genomic and routinely collected data [ 78 ].

Limitations

This review was not intended to be exhaustive or systematic; therefore, it does not include all health research studies that have used genomic and routinely collected data. We also excluded biobanks and databanks that do not house routine data; therefore, we only include examples here. This may have resulted in inadvertently excluding some countries and institutions where this research takes place and the types of genomic and routinely collected data that have been used. Not implementing systematic methods in study selection may have introduced bias in this review’s conclusions; however, the pragmatic approach used here was deemed sufficient to meet our objectives.

We included 19 studies in this review, only one of these involved data and research from a non-Western country (China) [ 21 ]. We did not come across any research that had taken place in low- to middle-income countries, although as this review was not conducted systematically, we may have unintentionally overlooked these. However, the absence of any studies from such countries may be, as Tekola-Ayele and Rotimi [ 79 ] explain, a result of having a limited number of well-trained genomic scientists and poor research infrastructure, and due to a less well-established routine data collection infrastructure and procedures such as EHRs [ 80 ].

Qualitative interviews may be subject to recruitment bias, which means that some viewpoints and experiences were excluded. In addition, there is limited information about participants; however, as this is a relatively small field of research, it was deemed necessary to maintain participants’ privacy.

Given the projected increase in the availability of genomic data, the potential to be obtained from its combination with routine health data is vast. This review has shown examples of what has been done in this field so far with, for example, GWAS and PheWAS plus other study designs. For fields such as pharmacogenomics, these methodologies need to be used further, where using routinely collected data will simplify the process of tracking longer-term outcomes of personalized medical treatments, and elucidate new findings on the effects of the environment on drug-gene interactions. Our take away from this study is that there are several challenges involved in using these data, particularly surrounding privacy. Therefore, it is imperative that appropriate data governance be documented and that public engagement activities take place to ensure socially acceptable practices. Overcoming these barriers will ensure that the use of these data to progress health research can be exploited to its full potential.

Acknowledgments

This study was funded by the UK Medical Research Council: MC_PC_16035.

Abbreviations

BCBritish Columbia
EHRelectronic health record
eMERGEElectronic and Medical Records and Genomics
GAPGenetics and Psychosis
GWASGenome-Wide Association Studies
ICDInternational Classification of Disease
NHSNational Health Service
PheWASphenome-wide association studies
PONDProvince of Ontario Neurodevelopmental Disorders
SAILSecure Anonymized Information Linkage

Multimedia Appendix 1

Conflicts of Interest: None declared.

  • Resource Library
  • Latest Topics
  • Ask a Question
  • Take a Tour
  • Recently Added
  • Featured Images
  • Featured Articles
  • Most Viewed Articles
  • Random Article
  • Add New Word
  • Random Term
  • Recent Changes
  • Share Your Resources

jeff is conducting a literature review on the research on genetics

  • Advanced Search
  • Image Gallery
   

Jeff is conducting a literature review on the research on genetics and sexual orientation. He would ...

Porkles wrote...

jeff is conducting a literature review on the research on genetics

Reply

Related Topics

  • Solved A literature review before conducting research serves which purpose? (Select all that apply.)1. It ...
  • Solved What are the steps to conducting a systematic literature review?
  • In a comprehensive review of research on the role of biology in determining sexual orientation, the ...
  • Conducting a literature review means:
  • Solved A nurse is very interested in conducting research on a specific topic. Review of the literature ...
  • Solved Which objective of a review of the literature is unique to a review for research purposes, rather ...
  • Solved In historical research, how does the literature review fit into the study?
  • Solved Which of the following sources would be most useful for a researcher conducting a literature review? ...
Replies wrote...
       
©

jeff is conducting a literature review on the research on genetics

  • Which of the following is the best approach to mitigate alignment risk? - Accounting
  • Parallel or concurrent tasks are tasks that be done at the same time. - Accounting
  • The analysis phase of the SDLC involves a complete, detailed analysis of the systems needs of the ... - Accounting
  • The benefits of an IT project are not necessarily measurable in financial terms. - Accounting
  • The 15-15 Rule states that if a project is more than 15 percent over budget or 15 percent off the ... - Accounting
  • The design phase of the SDLC begins with a business need for a new or better information system. - Accounting
  • The Sarbanes-Oxley Act's 404 Reports require management and auditors to report on - Accounting
  • Which of the following would not help mitigate change risk? - Accounting
  • The economic justification process for a new IT initiative includes all of the following except: - Accounting
  • Benefits are often estimated without complete information. - Accounting
Join Our Community
is a free online homework help service catered towards college and high school students. Get homework help and answers to your toughest questions in biology, chemistry, physics, mathematics, engineering, accounting, business, humanities, and more. Master your assignments quickly with thousands of step-by-step solutions to countless textbook questions asked and answered by our members. If we don't have your question, don't worry. You can ask any homework question and get expert homework help in as little as two hours.
Home Quick Links Social Facebook YouTube Help
| |
Biology-Forums.com is not affiliated with any publisher. Book covers, title and author names appear for reference only.

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Perspective
  • Published: 29 April 2024

Advancing genomics to improve health equity

  • Ebony B. Madden   ORCID: orcid.org/0000-0003-4510-9563 1 ,
  • Lucia A. Hindorff 1 ,
  • Vence L. Bonham 1 ,
  • Tabia Henry Akintobi 2 ,
  • Esteban G. Burchard   ORCID: orcid.org/0000-0001-7475-2035 3 ,
  • Kellan E. Baker 4 ,
  • Rene L. Begay 5 ,
  • John D. Carpten 6 ,
  • Nancy J. Cox 7 ,
  • Valentina Di Francesco   ORCID: orcid.org/0009-0001-9208-5560 1 ,
  • Denise A. Dillard 8 ,
  • Faith E. Fletcher 9 ,
  • Stephanie M. Fullerton   ORCID: orcid.org/0000-0002-0938-6048 10 ,
  • Nanibaa’ A. Garrison   ORCID: orcid.org/0000-0002-6228-3216 11 , 12 , 13 ,
  • Catherine M. Hammack-Aviran 14 ,
  • Vanessa Y. Hiratsuka   ORCID: orcid.org/0000-0002-4234-9441 15 ,
  • James E. K. Hildreth 16 ,
  • Carol R. Horowitz 17 ,
  • Chanita A. Hughes Halbert 18 ,
  • Michael Inouye   ORCID: orcid.org/0000-0001-9413-6520 19 ,
  • Amber Jackson 1 ,
  • Latrice G. Landry 20 ,
  • Rick A. Kittles 21 ,
  • Jeff T. Leek 22 ,
  • Nita A. Limdi 23 ,
  • Nicole C. Lockhart 1 ,
  • Elizabeth O. Ofili 24 ,
  • Eliseo J. Pérez-Stable 25 ,
  • Maya Sabatello 26 , 27 ,
  • Loren Saulsberry   ORCID: orcid.org/0000-0003-0325-2329 28 ,
  • Lorjetta E. Schools 29 ,
  • Jennifer L. Troyer 1 ,
  • Benjamin S. Wilfond 30 ,
  • Genevieve L. Wojcik   ORCID: orcid.org/0000-0001-7206-8088 31 ,
  • Judy H. Cho   ORCID: orcid.org/0000-0002-7959-0466 17 ,
  • Sandra S.-J. Lee 27 &
  • Eric D. Green 1  

Nature Genetics volume  56 ,  pages 752–757 ( 2024 ) Cite this article

2460 Accesses

32 Altmetric

Metrics details

  • Genetics research

Health equity is the state in which everyone has fair and just opportunities to attain their highest level of health. The field of human genomics has fallen short in increasing health equity, largely because the diversity of the human population has been inadequately reflected among participants of genomics research. This lack of diversity leads to disparities that can have scientific and clinical consequences. Achieving health equity related to genomics will require greater effort in addressing inequities within the field. As part of the commitment of the National Human Genome Research Institute (NHGRI) to advancing health equity, it convened experts in genomics and health equity research to make recommendations and performed a review of current literature to identify the landscape of gaps and opportunities at the interface between human genomics and health equity research. This Perspective describes these findings and examines health equity within the context of human genomics and genomic medicine.

This is a preview of subscription content, access via your institution

Access options

Access Nature and 54 other Nature Portfolio journals

Get Nature+, our best-value online-access subscription

24,99 € / 30 days

cancel any time

Subscribe to this journal

Receive 12 print issues and online access

195,33 € per year

only 16,28 € per issue

Buy this article

  • Purchase on Springer Link
  • Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

jeff is conducting a literature review on the research on genetics

Similar content being viewed by others

jeff is conducting a literature review on the research on genetics

Strategic vision for improving human health at The Forefront of Genomics

jeff is conducting a literature review on the research on genetics

A roadmap to increase diversity in genomic studies

jeff is conducting a literature review on the research on genetics

Genomic medicine implementation protocols in the PhenX Toolkit: tools for standardized data collection

Carnethon, M. R., Kershaw, K. N. & Kandula, N. R. Disparities research, disparities researchers, and health equity. JAMA 323 , 211–212 (2020).

Article   PubMed   Google Scholar  

Gomez, C. A. et al. Addressing health equity and social determinants of health through Healthy People 2030. J. Public Health Manag. Pract. 27 , S249–S257 (2021).

Article   PubMed   PubMed Central   Google Scholar  

Westfall, J. M., Roper, R., Gaglioti, A. & Nease, D. E. Jr. Practice-based research networks: strategic opportunities to advance implementation research for health equity. Ethn. Dis. 29 , 113–118 (2019).

Google Scholar  

Jooma, S., Hahn, M. J., Hindorff, L. A. & Bonham, V. L. Defining and achieving health equity in genomic medicine. Ethn. Dis. 29 , 173–178 (2019).

Bonham, V. L. Examining how race, ethnicity, and ancestry data are used in biomedical research. JAMA 320 , 1533–1534 (2018).

Saunkeah, B., Beans, J. A., Peercy, M. T., Hiratsuka, V. Y. & Spicer, P. Extending research protections to tribal communities. Am. J. Bioeth. 21 , 5–12 (2021).

McDonald, K. E., Schwartz, A. E. & Sabatello, M. Eligibility criteria in NIH-funded clinical trials: can adults with intellectual disability get in? Disabil. Health J. 15 , 101368 (2022).

Dickson, E. et al. Characteristics and practices within research partnerships for health and social equity. Nurs. Res. 69 , 51–61 (2020).

Castillo, E. G. & Harris, C. Directing research toward health equity: a health equity research impact assessment. J. Gen. Intern. Med. 36 , 2803–2808 (2021).

Horowitz, C. R. et al. Race, genomics and chronic disease: what patients with African ancestry have to say. J. Health Care Poor Underserved 28 , 248–260 (2017).

O’Daniel, J. M. et al. Integration of stakeholder engagement from development to dissemination in genomic medicine research: approaches and outcomes from the CSER Consortium. Genet. Med . 24 , 1108–1119 (2022).

Liburd, L. C. et al. Addressing health equity in public health practice: frameworks, promising strategies, and measurement considerations. Annu. Rev. Public Health 41 , 417–432 (2020).

Krieger, N. Measures of racism, sexism, heterosexism, and gender binarism for health equity research: from structural injustice to embodied harm—an ecosocial analysis. Annu. Rev. Public Health 41 , 37–62 (2020).

Dover, D. C. & Belon, A. P. The health equity measurement framework: a comprehensive model to measure social inequities in health. Int. J. Equity Health 18 , 36 (2019).

Penman-Aguilar, A. et al. Measurement of health disparities, health inequities, and social determinants of health to support the advancement of health equity. J. Public Health Manag. Pract. 22 , S33–S42 (2016).

Braveman, P. et al. What is health equity? Behav. Sci. Policy 4 , 1–14 (2018).

Article   Google Scholar  

Byeon, Y. J. J. et al. Evolving use of ancestry, ethnicity, and race in genetics research—a survey spanning seven decades. Am. J. Hum. Genet. 108 , 2215–2223 (2021).

Article   CAS   PubMed   PubMed Central   Google Scholar  

Mulder, N. J. et al. Development of bioinformatics infrastructure for genomics research. Glob. Heart . 12 , 91–98 (2017).

Download references

Acknowledgements

We thank the attendees of the Future Directions in Genomics and Health Equity Workshop, the NHGRI Training, Diversity and Health Equity Office and the NHGRI Office of Communication for their expertise, assistance and thoughtful perspectives and recommendations.

Author information

Authors and affiliations.

Office of the Director, National Human Genome Research Institute, Bethesda, MD, USA

Ebony B. Madden, Lucia A. Hindorff, Vence L. Bonham, Valentina Di Francesco, Amber Jackson, Nicole C. Lockhart, Jennifer L. Troyer & Eric D. Green

Department of Community Health and Preventative Medicine, Morehouse School of Medicine, Atlanta, GA, USA

Tabia Henry Akintobi

Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA

Esteban G. Burchard

Whitman-Walker Institute, Washington, DC, USA

Kellan E. Baker

Centers for American Indian and Alaska Native Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA

Rene L. Begay

Comprehensive Cancer Center, City of Hope, Duarte, CA, USA

John D. Carpten

Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA

Nancy J. Cox

Department of Medical Education and Clinical Sciences, Washington State University College of Medicine, Seattle, WA, USA

Denise A. Dillard

Center for Medical Ethics and Health Policy, Baylor College of Medicine, Houston, TX, USA

Faith E. Fletcher

Department of Bioethics and Humanities, University of Washington, Seattle, WA, USA

Stephanie M. Fullerton

Institute for Society and Genetics, University of California, Los Angeles, Los Angeles, CA, USA

Nanibaa’ A. Garrison

Institute for Precision Health, UCLA David Geffen School of Medicine, Los Angeles, CA, USA

Division of General Internal Medicine and Health Services Research, UCLA David Geffen School of Medicine, Los Angeles, CA, USA

Department of Health Policy, Vanderbilt University, Nashville, TN, USA

Catherine M. Hammack-Aviran

Center for Human Development, University of Alaska Anchorage, Anchorage, AK, USA

Vanessa Y. Hiratsuka

Meharry Medical College, Nashville, TN, USA

James E. K. Hildreth

Icahn School of Medicine at Mount Sinai, New York, NY, USA

Carol R. Horowitz & Judy H. Cho

Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA

Chanita A. Hughes Halbert

Department of Public Health and Primary Care, University of Cambridge Victor Phillip Dahdaleh Heart and Lung Research Institute, Cambridge, UK

Michael Inouye

Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA

Latrice G. Landry

Morehouse School of Medicine, Atlanta, GA, USA

Rick A. Kittles

Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, USA

Jeff T. Leek

Department of Neurology, University of Alabama, Birmingham School of Medicine, Birmingham, AL, USA

Nita A. Limdi

Department of Medicine, Morehouse School of Medicine, Atlanta, GA, USA

Elizabeth O. Ofili

National Institute on Minority Health and Health Disparities, Bethesda, MD, USA

Eliseo J. Pérez-Stable

Center for Precision Medicine and Genomics, Columbia University, New York, NY, USA

Maya Sabatello

Department of Medical Humanities and Ethics, Columbia University, New York, NY, USA

Maya Sabatello & Sandra S.-J. Lee

Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA

Loren Saulsberry

Common Fund, National Institutes of Health, Bethesda, MD, USA

Lorjetta E. Schools

Department of Pediatrics, University of Washington, Seattle, WA, USA

Benjamin S. Wilfond

Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA

Genevieve L. Wojcik

You can also search for this author in PubMed   Google Scholar

Contributions

E.B.M. took the lead in writing the manuscript with input from all authors. V.L.B., E.D.G., E.J.P.-S., L.A.H. and S.S.-J.L. made major contributions and performed critical revisions to the manuscript. R.L.B., N.C.L. and M.S. have given critical feedback and edits to the manuscript. T.H.A., K.E.B., E.G.B., J.D.C., N.J.C., V.D.F., D.A.D., F.E.F., S.M.F., N.A.G., V.Y.H., C.M.H.-A., J.E.H., C.R.H., C.A.H.H., M.I., A.J., R.A.K., L.G.L., J.T.L., N.A.L., E.O.O., L.S., L.E.S., J.L.T., B.S.W., G.L.W. and J.H.C. made content recommendations that helped shape the manuscript. All authors provided critical feedback and helped shape the manuscript.

Positionality statement:

R.L.B. and N.A.G. are members of the Diné tribe. D.A.D. is a member of the Eskimo tribe. V.Y.H. is a member of the Navajo and Wintu tribes.

Corresponding author

Correspondence to Ebony B. Madden .

Ethics declarations

Competing interests.

M.S. is a member of the Institutional Review Board for the All of Us Research Program. M.I. is a trustee of the Public Health Genomics Foundation, is a member of the Scientific Advisory Board of Open Targets and has research collaborations with AstraZeneca, Nightingale Health and Pfizer (unrelated to the work of this article). All other authors confirm that there are no financial or nonfinancial interests that may bias the conclusions, implications or opinions stated.

Peer review

Peer review information.

Nature Genetics thanks Alice Popejoy and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Cite this article.

Madden, E.B., Hindorff, L.A., Bonham, V.L. et al. Advancing genomics to improve health equity. Nat Genet 56 , 752–757 (2024). https://doi.org/10.1038/s41588-024-01711-z

Download citation

Received : 20 June 2023

Accepted : 08 March 2024

Published : 29 April 2024

Issue Date : May 2024

DOI : https://doi.org/10.1038/s41588-024-01711-z

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

jeff is conducting a literature review on the research on genetics

IMAGES

  1. basic steps of conducting literature review

    jeff is conducting a literature review on the research on genetics

  2. How To Write A Literature Review Research

    jeff is conducting a literature review on the research on genetics

  3. Practical Strategies and Guidelines for Conducting Literature Reviews

    jeff is conducting a literature review on the research on genetics

  4. basic steps of conducting literature review

    jeff is conducting a literature review on the research on genetics

  5. The Importance of Literature Review in Scientific Research Writing

    jeff is conducting a literature review on the research on genetics

  6. Systematic Literature Review Methodology

    jeff is conducting a literature review on the research on genetics

COMMENTS

  1. Chapter 11 Reading Assessment Flashcards

    Jeff is conducting a literature review on the research on genetics and sexual orientation. He would find which of the following statements are supported by research? Over the past 30 years, research methods have become more sophisticated, allowing researchers to tease apart the forces of environment and genes.

  2. Steps in Conducting a Literature Review

    A literature review is important because it: Explains the background of research on a topic. Demonstrates why a topic is significant to a subject area. Discovers relationships between research studies/ideas. Identifies major themes, concepts, and researchers on a topic. Identifies critical gaps and points of disagreement.

  3. A theory-informed systematic review of clinicians' genetic testing

    This systematic literature review investigates factors impacting on clinicians' decisions to offer genetic tests in their practice, and maps them to a theoretical behaviour change framework.

  4. The road ahead in genetics and genomics

    Abstract. In celebration of the 20th anniversary of Nature Reviews Genetics, we asked 12 leading researchers to reflect on the key challenges and opportunities faced by the field of genetics and ...

  5. How is genetic testing evaluated? A systematic review of the literature

    Given the rapid development of genetic tests, an assessment of their benefits, risks, and limitations is crucial for public health practice. We performed a systematic review aimed at identifying and comparing the existing evaluation frameworks for genetic tests. We searched PUBMED, SCOPUS, ISI Web of Knowledge, Google Scholar, Google, and gray ...

  6. How is genetic testing evaluated? A systematic review of the literature

    Discussion. Our review identified three main approaches to the evaluation of genetic testing: the ACCE model, the HTA process, and the Wilson and Jungner screening criteria. The most popular is the ACCE model, developed in 2000 by the US Centers for Disease Control and Prevention [ 7 ].

  7. How is genetic testing evaluated? A systematic review of the literature

    Given the rapid development of genetic tests, an assessment of their benefits, risks, and limitations is crucial for public health practice. We performed a systematic review aimed at identifying ...

  8. Hereditary Cancer Genetic Panel Testing: A Review of the Literature

    The author conducted a literature review to outline the science on CGT methods, the psychosocial responses to testing among patients, and the unique role of nurses in this process. A significant gap in the literature exists regarding multigene cancer genetic panel tests and the associated experiences and decision-making processes among ...

  9. (PDF) Definition and conceptual model of genetics literacy: a

    Genetic literacy is primarily defined as sufficient knowledge and appreciation of genetics. principles to allow informed decision-making for personal well -being and effective participation in ...

  10. What do people think about genetics? A systematic review

    The present study reviews the current literature on what people in the US report about genetics: their attitudes, knowledge, awareness, and perceptions. This review provides the background setting needed to assist the development of community-specific and population-level assessment and engagement tools to increase community participation in ...

  11. Predictors of Genetic Testing Decisions: A Systematic Review and

    Of note, we opted to conduct a qualitative systematic review rather than a quantitative (i.e., meta-analytic) review. Our ability to discuss the wide array of considerations that influence genetic testing decisions, many of which rely on statistical procedures that are difficult or impossible to properly synthesize (e.g., multiple regression, structural equation modeling), would be limited by ...

  12. Exploring the Use of Genomic and Routinely Collected Data: Narrative

    Objective. This study aims to provide an outline of the use of genomic data alongside routinely collected data in health research to date. As this field prepares to move forward, it is important to take stock of the current state of play in order to highlight new avenues for development, identify challenges, and ensure that adequate data governance models are in place for safe and socially ...

  13. Jeff is conducting a literature review on the research on genetics and

    Our community brings together students, educators, and subject enthusiasts in an online study community. With around-the-clock expert help, you can find the help you need, whenever you need it.

  14. Parenting is genetically influenced: What does that mean for research

    1.4 Methodological challenges and limitations of genetically-informed research on parenting. Notwithstanding the advantages of genetically-informed research, social science genetics is very much a field in development, and studies warrant replication before findings should be used as input for practical recommendations.

  15. Direct-to-consumer genomic testing: systematic review of the literature

    Conducting a systematic review is a way of identifying evidence of adequate quality about a specific topic and synthesising it in a way that is rigorous and transparent. 12 In this study, we used ...

  16. The Nature-Nurture Question #8 Flashcards

    Study with Quizlet and memorize flashcards containing terms like As a general statement, the higher the ______ coefficient, the stronger the influence of one's genes on a specific trait being examined. correlation genetic ethnographic heritability biological, Denzel is a psychologist who is interested in conducting research that examines the nature-nurture debate as it applies to people. He is ...

  17. SOCIAL PSYCH: CH.1 QUIZ 1 Flashcards

    SOCIAL PSYCH: CH.1 QUIZ 1. Jeff is a social psychologist who favors the "cold" approach to understanding human behavior. He is conducting research on why people fail to use condoms even when they know that condoms can reduce their risk of contracting HIV. Jeff is least likely to devote his attention to people's.

  18. The design, implementation, and effectiveness of intervention ...

    Despite rapid advancements in genetics and genomics, referral practices remain suboptimal. This systematic review assesses the extent to which approaches from implementation science have been ...

  19. Chapter 11 Flashcards

    Study with Quizlet and memorize flashcards containing terms like Which of the following statements is true about sexual orientation and protected classes in discrimination law?, Jane identifies as a lesbian. If Jane's college experience is like that of many other gay, lesbian, and bisexual students, which of the following statements would be least likely to be true?, In the past, all states in ...

  20. PDF How is genetic testing evaluated? A systematic review of the literature

    Given the rapid development of genetic tests, an assessment of their benefits, risks, and limitations is crucial for public health practice. We performed a systematic review aimed at identifying ...

  21. HDFS 312 Exam 1 Quiz Questions Flashcards

    Study with Quizlet and memorize flashcards containing terms like Dr. Cundy, a behavior geneticist, is interested in conducting a twin study to investigate the influence genetics may have on a person's propensity to be married. What problem is Dr. Cundy likely to face?, Carly was a teenage mom, who then became a grandmother at 35. Carly also found out that she was pregnant at the same time as ...

  22. Advancing genomics to improve health equity

    The consensus of both the workshop and the literature review is that research is needed to understand both the effects of genomics on health equity and how to disseminate and implement genomic ...

  23. HDFS 276 Exam 1 Flashcards

    Study with Quizlet and memorize flashcards containing terms like The sensation stage of the Erotic Stimulus Pathway Theory is similar to the _____ and _____ phases of the EPOR model., How does the research of Levin differ from Kaplan's model of sexual arousal?, Abdul is experiencing the sensation of ejaculatory inevitability. This means that _____. and more.