Skip to main content
Log in

A comprehensive genomic reporting structure for communicating all clinically significant primary and secondary findings

  • Original Investigation
  • Published:
Human Genetics Aims and scope Submit manuscript

This article has been updated

Abstract

Genomic sequencing (GS) can reveal secondary findings (SFs), findings unrelated to the reason for testing, that can be overwhelming to both patients and providers. An effective approach for communicating all clinically significant primary and secondary GS results is needed to effectively manage this large volume of results. The aim of this study was to develop a comprehensive approach to communicate all clinically significant primary and SF results. A genomic test report with accompanying patient and provider letters were developed in three phases: review of current clinical reporting practices, consulting with genetic and non-genetics experts, and iterative refinement through circulation to key stakeholders. The genomic test report and consultation letters present a myriad of clinically relevant GS results in distinct, tabulated sections, including primary (cancer) and secondary findings, with in-depth details of each finding generated from exome sequencing. They provide detailed variant and disease information, personal and familial risk assessments, clinical management details, and additional resources to help support providers and patients with implementing healthcare recommendations related to their GS results. The report and consultation letters represent a comprehensive approach to communicate all clinically significant SFs to patients and providers, facilitating clinical management of GS results.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Data availability

The data and findings in this study are available in the manuscript and/or supplemental material.

Change history

  • 08 September 2022

    The article is updated to correct the acknowledgement.

References

Download references

Acknowledgements

This study was supported by a Foundation Grant from the Canadian Institutes of Health Research and a Quality of Life Grant from the Canadian Cancer Society Research Institute awarded to Yvonne Bombard (Grant #s 143310 and 705665, respectively). Yvonne Bombard was supported by a New Investigator Award from the Canadian Institute of Health Research (Grant #136664) during the conduct of this study. Jordan Lerner-Ellis was funded by the McLaughlin Centre (Grant #MC-2012-13 and #MC-2014-11-1) and CIHR-Champions of Genetics: Building the Next Generation Grant (FRN: 135730).

Incidental Genomics study team: Yvonne Bombard1,2,9, Susan Randall Armel2,7, Melyssa Aronson3, Nancy Baxter12,13,14,20, Kenneth Bond15, José-Mario Capo-Chichi4, June C. Carroll2,3, Timothy Caulfield16,17,18, Marc Clausen1, Tammy J. Clifford19, Iris Cohn5, Irfan Dhalla12,20,21,22,23, Craig C. Earle22, Andrea Eisen6, Christine Elser2,3, Michael Evans12, Emily Glogowski24, Tracy Graham2,6, Elena Greenfeld25,26, Jada G. Hamilton27, Wanrudee Isaranuwatchai12,20, Monika Kastner12,20,28, Raymond H. Kim2,3,4,5, Andreas Laupacis12,20, Jordan Lerner-Ellis2,3,10,11, Chantal F. Morel4, Michelle Mujoomdar29, Abdul Noor25,26, Kenneth Offit27, Seema Panchal3, Mark E. Robson27, Stephen W. Scherer5,22,30, Adena Scheer12,13, Kasmintan A. Schrader8, Terrence Sullivan20 and Kevin E. Thorpe31,32.

12St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada. 13Department of Surgery, University of Toronto, Toronto, Canada. 14Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia. 15Institute of Health Economics, Edmonton, AB, Canada. 16Faculty of Law, University of Alberta, Edmonton, AB, Canada. 17School of Public Health, University of Alberta, Edmonton, AB, Canada. 18Health Law Institute, University of Alberta, Edmonton, AB, Canada. 19School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada. 20Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada. 21Institute for Clinical Evaluative Sciences, Toronto, ON, Canada. 22Department of Medicine, University of Toronto, Toronto, ON, Canada. 23Health Quality Ontario, Toronto, ON, Canada. 24GeneDx, Gaithersburg, MD, USA. 25Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada. 26Division of Diagnostic Medical Genetics, Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada. 27Memorial Sloan Kettering Cancer Center, New York, NY, USA. 28Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada. 29Canadian Agency for Drugs and Technologies in Health, Ottawa, ON, Canada. 30Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada. 31Applied Health Research Centre (AHRC), Li Ka Shing Knowledge Institute of St. Michael’s Hospital, Toronto, ON, Canada. 32Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada.

Funding

This study was supported by a Foundation Grant from the Canadian Institutes of Health Research and a Quality of Life Grant from the Canadian Cancer Society Research Institute (grant numbers 143310 and 705665, respectively).

Author information

Authors and Affiliations

Authors

Consortia

Contributions

JS, ER, RK, AS, MGS, SS, YB, and JLE conceptualized the study. Formal analysis was performed by JS, ER, RK, MGS, SS, JLE, and YB. Investigation was performed by JS, ER, AS, RK, MGS, and SS. This study was supervised by YB and JLE. JS was involved in visualization. The original draft was written by JS, ER, RK, and AS. Manuscript review and editing involved JS, ER, RK, YB, JLE, JCC, SRA, AS, MC, MGS, SS, CM, MA, JMCC, IC, AE, CE, TG, KO, SP, CP, KAS, and RHK. All the authors have approved the final draft submitted.

Corresponding authors

Correspondence to Jordan Lerner-Ellis or Yvonne Bombard.

Ethics declarations

Conflict of interest

On behalf of all the authors, the corresponding author states that there is no conflict of interest.

Ethical approval

This study was approved by the Research Ethics Board of Unity Health Toronto–St.Michael’s Hospital (CTO0819); ClinicalTrial.gov identifier NCT03597165. Informed consent was obtained from all the participants involved in this study. Any individual-level data, including clinical data, found in this manuscript or supplementary files have been de-identified and pseudonyms were used where appropriate.

Additional information

Publisher's Note

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

The article is updated to correct the acknowledgement.

Supplementary Information

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sam, J., Reble, E., Kodida, R. et al. A comprehensive genomic reporting structure for communicating all clinically significant primary and secondary findings. Hum Genet 141, 1875–1885 (2022). https://doi.org/10.1007/s00439-022-02466-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00439-022-02466-5

Navigation