American Journal of Medical Genetics Part C (Seminars in Medical Genetics) 166C:1–7 (2014)

I N V I T E D C O M M E N T

Leading the Way to Genomic Medicine TERI A. MANOLIO*

AND

ERIC D. GREEN

The National Human Genome Research Institute, in close collaboration with its research community, is pursuing an ambitious research agenda to facilitate and promote the implementation of genomics in clinical care. Since 2011, research programs utilizing next‐generation sequencing in the management of cancer and other multigenic conditions, workup of undiagnosed conditions, and evaluation of disorders of the newborn period have been initiated, along with projects identifying clinically actionable variants and exploring the ethical and social implications of reporting these findings. Several genomic medicine symposia and other consultations have helped to shape these research initiatives and develop educational materials for physicians and others working to implement the use of genomic findings in clinical care. These efforts provide a valuable complement to the highly successful basic genomics research enterprise that has at last enabled the transition of genomics from the bench to the bedside. © 2014 Wiley Periodicals, Inc.

How to cite this article: Manolio TA, Green ED. 2014. Leading the way to genomic medicine. Am J Med Genet Part C Semin Med Genet 166C:1–7.

INTRODUCTION In 2011, the National Human Genome Research Institute (NHGRI) published an updated strategic vision for the future of genomics research [Green and Guyer, 2011], proposing an ambitious research agenda that would facilitate and promote the implementation of genomics in clinical care (i.e., “genomic medicine”). This vision is framed around five research domains, three of which reflect fundamental technologic and basic science pursuits that together will advance our understanding of the role of the human genome in health and disease. While these three research domains—investigating genome structure, genome biology, and the (genomic) biology of disease—have long been (and will continue to be) key components of the Institute’s core mission, the 2011 strategic vision described two additional domains focused on genomics research aiming to advance the science of medicine and to improve the effectiveness of

healthcare. Together, these last two research domains represent a concerted effort to apply genomics for improving the prevention, diagnosis, and treatment of human disease.

RESEARCH NEEDS AND GENOMIC MEDICINE CIRCA 2011 Although a handful of genomic applications were already in clinical practice in 2011 (e.g., the use of specific tumor mutations in cancer treatment and HLA testing prior to abacavir use [Green and Guyer, 2011]), it was incontrovertibly clear that considerable research would be needed to capitalize on new genomic discoveries for implementing genomic medicine. Among the needed studies were those demonstrating the generalizability of genomic findings across populations and clinical settings (as well as potential interactions of genomic variants with other conditions or treat-

Although a handful of genomic applications were already in clinical practice in 2011 (e.g., the use of specific tumor mutations in cancer treatment and HLA testing prior to abacavir use [Green and Guyer, 2011]), it was incontrovertibly clear that considerable research would be needed to capitalize on new genomic discoveries for implementing genomic medicine. ments) and those generating evidence of the efficacy of using genomic information for clinical care. Such studies

Dr. Teri A. Manolio, M.D., Ph.D. is a Physician‐Epidemiologist who directs the Division of Genomic Medicine of the National Human Genome Research Institute of the National Institutes of Health. Dr. Eric D. Green is a Physician‐Scientist who directs the National Human Genome Research Institute of the National Institutes of Health. *Correspondence to: Teri A. Manolio, M.D., Ph.D., Director, Division of Genomic Medicine, National Human Genome Research Institute, 5635 Fishers Lane, Room 4113, MSC 9305, Bethesda, MD 20892‐9305. E‐mail: [email protected] DOI 10.1002/ajmg.c.31384 Article first published online in Wiley Online Library (wileyonlinelibrary.com): 11 March 2014

ß 2014 Wiley Periodicals, Inc.

2

AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS)

typically require vast numbers of patients to identify those with particular genomic variants and conditions of interest; this can be done efficiently by leveraging established biorepositories that are linked with electronic medical records (EMRs). It is notable that the latter situation is also ideal for rapidly acting on genomic findings once they are confirmed. By 2011, the value of using EMRs for assessing phenotypes (“electronic phenotyping”) had been demonstrated [Denny et al., 2010; Ritchie et al., 2010], but many questions remained about integrating genomic information into EMRs, maintaining patient privacy, and providing computerized decision support for practicing healthcare professionals. Also by 2011, rapid advances in “next‐generation” DNA sequencing technologies were being applied to difficult clinical problems, such as uncovering mutations underlying Mendelian syndromes [Ng et al., 2010; Lindhurst et al., 2011] and optimizing management of patients with rare disorders [Bainbridge et al., 2011]. Exome and genome sequencing was beginning to be used in the workup of patients with undiagnosed conditions [Worthey et al., 2011; St. Hilaire et al., 2011], but the feasibility of such approaches outside of highly specialized centers and their generalizability to broader categories of patients remained unclear. Such extensive genomic characterization also raised challenging issues relating to data sharing, informed consent, and the reporting of incidental genomic findings unrelated to the index condition but with potential implications for clinical care. Finally, the potential of genome sequencing to augment or even replace the standard approaches to screen for hereditary diseases in newborns raised numerous questions about efficacy, feasibility, and psychosocial impact that had yet to be fully formulated, let alone addressed [National Human Genome Research Institute, 2010]. At the same time, genomic analyses of tumors (by genotyping and/or sequencing methods) were becoming relatively common in the clinical setting [Dias‐Santagata et al., 2010], but few

medical centers were otherwise actively engaged in implementing genomic medicine. Some early clinical genomic successes were occurring in isolation, seemingly without strong means to disseminate their expertise or to develop common knowledgebases and other infrastructure [Manolio et al., 2013]. There appeared to be considerable motivation to develop implementation strategies that built upon the lessons learned by the early adopters, yet a robust forum for facilitating such development and dissemination was lacking. Meanwhile, most physicians and other healthcare professionals were largely unaware of genomic advances that might be relevant to their patients and were generally intimidated by the rapidly emerging discipline of genomic medicine, with few feeling competent to use genomics in their practice [Feero and Green, 2011].

Most physicians and other healthcare professionals were largely unaware of genomic advances that might be relevant to their patients and were generally intimidated by the rapidly emerging discipline of genomic medicine, with few feeling competent to use genomics in their practice [Feero and Green, 2011].

NHGRI CONSULTATIONS AND MEETINGS ON GENOMIC MEDICINE Shortly after publishing its 2011 strategic vision, NHGRI embarked on a series of consultations with other NIH Institutes and Centers to identify genomic medicine projects related to disease prevention, diagnosis, or treatment that were nearly ready for implementation. Despite great enthusiasm for clinical appli-

INVITED COMMENT

cations of genomics, most Institutes/ Centers identified projects falling more in the realm of NHGRI’s “biology of disease” domain (Table I). Relatively few projects involved examining the impact of using individual patients’ genomic information in their medical care (NHGRI’s “science of medicine” domain), and almost none focused on demonstrating the utility of genomics for actually improving the care of patients (NHGRI’s “effectiveness of healthcare” domain). This fifth domain encompasses much of what is referred to as “implementation research”—the study of methods that promote the systematic uptake of proven interventions into routine clinical care [Eccles et al., 2012]. Of note, one project in this realm supported early on by NHGRI involved the targeted sequencing of 84 pharmacologically important genes by the Electronic Medical Records and Genomics (eMERGE) network (see below). In parallel, NHGRI also consulted and facilitated collaborations with the external research community by convening a series of “Genomic Medicine Meetings” (Table II), developed with the guidance and leadership of the Genomic Medicine Working Group of the National Advisory Council on Human Genome Research [National Human Genome Research Institute a]. The first meeting in June 2011 laid to rest any doubts about whether there existed a critical mass of researchers actively engaged in genomic medicine implementation; representatives of 20 groups attended this first meeting on short notice and on their own funds. Commonalities and duplications across efforts became readily apparent, including similar obstacles encountered and solutions developed, often quite independently. A summary of these efforts and the major lessons learned by early adopters was published as an “implementation roadmap” [Manolio et al., 2013], and plans were made to facilitate collaborations and to address the critical need for a consensus process to identify clinically actionable genomic variants. Additional meetings in December 2011 addressed the latter two topics,

INVITED COMMENT

AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS)

3

TABLE I. Disease‐Related Genomics Research Across Three NHGRI Research Domains NHGRI domain

Biology of disease

Common Rubric General Goal

Discovery research Demonstrate genotype– phenotype associations

Specific Examples

Identify persons at increased risk of disease based on their genomic variants Find all variants related to given phenotype or disease

Characterize variation in genes known to be related to disease or treatment response Characterize phenotypic variation and effect modifiers in carriers of specific variants

Classify patients into subgroups with differing prognosis or treatment response (molecular taxonomy)

leading to the release of several NHGRI funding solicitations and ultimately the funding of two new consortia, the Clinical Genomics Resource (ClinGen) [Ramos et al., 2014] and Implementing Genomics into Clinical Practice (IGNITE) Network (see below). Later meetings focused on issues relevant to laboratories and payers (May 2012), professional societies (January 2013), and federal agencies (May 2013). Each of these led to follow‐up discussions regarding potential collaborative research projects with payers and/or with multiple federal healthcare providers. The fourth meeting was particularly productive, with the professional societies urging NHGRI to establish and co‐lead an Inter‐Society Coordinating

Science of medicine

Effectiveness of healthcare

Clinical validation Assess outcomes after using genomic information to direct clinical care Evaluate clinician and patient satisfaction with care after receipt of genomic information Assess impact of reporting incidental findings on health behaviors, healthcare utilization, and psychological well‐being Identify causes of rare or undiagnosed diseases

Clinical implementation Demonstrate improved healthcare with the use of genomic information Educate clinicians and patients in clinical use of genomic information Develop clinical informatics systems for reporting results of genomic analyses and providing decision support

Identify sources and susceptibilities of infectious agents

Compare genome sequencing to enzymatic and other assays for modifiable metabolic disorders in newborns Reclassify intermediate risk patients into high‐ and low‐risk categories where differential interventions available Validate drug targets and develop improved therapeutic agents

Committee on Practitioner Education in Genomics (ISCC) that would facilitate the efforts of professional societies in developing and sharing genomics educational materials and standards for physicians and other health practitioners [Manolio et al., 2013; Manolio and Murray, in press; National Human Genome Research Institute b]. The ISCC has already empanelled several working groups to develop competencies, use cases, and accessible educational products for use across multiple disciplines and professional organizations. A sixth genomic medicine meeting to explore potential international collaborations in genomic medicine is planned for January 2014. Subsequent meetings focusing on other key stakeholder

Evaluate impact of using genomic variant information to individualize treatment Incorporate genomic information into electronic medical records that can follow patients across care systems and throughout lifespan Define clinically actionable genomic variants and disseminate that information and its relevant evidence base

groups and topics are anticipated to be held roughly every 9 months. Other tangible products of these meetings, such as a potential large collaborative project across multiple federal healthcare providers, are currently in development.

NHGRI PROGRAMS IN GENOMIC MEDICINE Three research programs already in progress in 2011 readily lent themselves to extension into genomic medicine implementation studies: NHGRI’s Electronic Medical Records and Genomics (eMERGE) network [McCarty et al., 2011], NHGRI’s genome sequencing program [National Human Genome Research Institute c], and the NIH

4

AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS)

INVITED COMMENT

TABLE II. NHGRI Genomic Medicine Meetings and Related Workshops Meeting

Dates

Emphasis

Products

Meeting URL https://www. genome.gov/ 27547270 http://www. genome.gov/ 27546546

Genomic Medicine Symposium [Manolio et al., 2013] ClinAction Workshop

June 29, 2011

Academic medical centers

Implementation roadmap [Manolio et al., 2013], ClinAction workshop

December 2–3, 2011

Methods for identifying clinically actionable variants

Genomic Medicine II

December 5–6, 2011

Pilot demonstration projects

Genomic Medicine III

May 3–4, 2012

Payers’ meeting

October 24, 2012

Working with laboratories and payers Potential for collaborative research

RFA HG‐12‐016, Clinically Relevant Genetic Variants Resource; ClinGen Consortium [Ramos et al., 2014] RFAs HG‐12‐006 and ‐007, Genomic Medicine Pilot Demonstration Projects; IGNITE Consortium Payers’ meeting

Genomic Medicine IV

January 28–29, 2013

Physician education in genomics

Genomic Medicine V

May 28–29, 2013

Working with federal stakeholders

Inter‐Society Coordinating Committee

September 19–20, 2013

Physician competencies, use cases, educational materials, and collaborations with specialty boards

Genomic Medicine VI

January 8–9, 2014

Genomic Medicine VII (proposed)

September, 2014

Developing international collaborations Working with industry?

Undiagnosed Diseases Program [Gahl et al., 2012]. The eMERGE network was established in 2007 to explore how best to use biorepositories linked with EMRs in genomics research projects.

Physician education workgroup is preparing a summary of lessons learned about the problems encountered by clinicians in the pre‐ and post‐analytic phases of genetic test ordering Inter‐Society Coordinating Committee for Practitioner Education in Genomics (ISCC), [National Human Genome Research Institute b], approval for AJMG Seminars issue devoted to Implementation of Genomic Medicine Exploratory implementation project in collaboration with VA and military medical services Resources developed by ISCC will be posted on NHGRI Genetics and Genomics Competency Center (G2C2) and NHGRI Global Genetics and Genomics Community (G3C) websites; White Paper [Manolio and Murray, in press].

Having demonstrated the research value of such biorepositories [Denny et al., 2010; Ritchie et al., 2010], eMERGE was poised to explore the use of genomic information for clinical

https://www. genome.gov/ 27546373 https://www. genome.gov/ 27548693

https://www. genome.gov/ 27552294

https://www. genome.gov/ 27553865 http://www. g‐2‐c‐2.org/ index.php and http://www. g‐3‐c.org/en/ http://www. genome.gov/ 27555775

care as it prepared for its second phase [National Human Genome Research Institute d]. Ten projects were funded in 2011–2012 to continue eMERGE’s efforts in electronic phenotyping and

INVITED COMMENT

The eMERGE network was established in 2007 to explore how best to use biorepositories linked with EMRs in genomics research projects. Having demonstrated the research value of such biorepositories [Denny et al., 2010; Ritchie et al., 2010], eMERGE was poised to explore the use of genomic information for clinical care as it prepared for its second phase [National Human Genome Research Institute d].

genomic discovery research, while also expanding into genomic medicine implementation studies. These projects included pilot studies examining genome sequencing in the workup of undiagnosed diseases, the effects of returning high‐risk CFH, HFE, and FVL variants on physician and patient behaviors, and genomic versus clinical risk assessments in managing coronary disease and hypertensive nephropathy. As noted above, in 2012 eMERGE proposed initiating a collaborative project in pharmacogenomics with the National Institute of General Medical Sciences. The resulting eMERGE‐PGx program involves assessing 9,000 patients for genomic variants in 84 genes known to be important in the response to drug therapy through the Pharmacogenomics Research Network [2013]. A select number of Clinical Laboratory Improvement Act (CLIA)‐validated variants with well‐established clinical implications— as defined by programs such as the Clinical Pharmacogenetics Implementation Consortium (CPIC [Relling and Klein, 2011])—are being reported to clinicians and integrated into these patients’ EMRs with appropriate clinical

AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS)

decision support to assess the impact of this genomic information on clinical decision‐making. Established in 2008, the NIH Undiagnosed Diseases Program (UDP) seeks to establish diagnoses for patients who remain undiagnosed after exhaustive medical workups and to discover new disorders and insights into disease mechanisms [National Institutes of Health a]. By 2011, the UDP had established diagnoses in nearly a quarter of the evaluated patients and identified several new disorders, but

In 2008, the NIH Undiagnosed Diseases Program (UDP) seeks to establish diagnoses for patients who remain undiagnosed after exhaustive medical workups and to discover new disorders and insights into disease mechanisms [Pharmacogenomics Research Network, 2013]. By 2011, the UDP had established diagnoses in nearly a quarter of the evaluated patients and identified several new disorders, but the transferability of the program and expertise outside the unique setting the NIH Clinical Center and Intramural Research Program was unclear.

the transferability of the program and expertise outside the unique setting the NIH Clinical Center and Intramural Research Program was unclear. NHGRI has since worked with the other NIH Institutes and Centers

5

through the NIH Common Fund to expand the UDP into a multi‐center Undiagnosed Diseases Network, which will involve the establishment of several new clinical sites and core facilities [National Institutes of Health b]. Funding for this program should begin in early Fiscal Year 2014. The potential for clinicians to utilize genome sequence data for the care of their patients is being explored by the Clinical Sequencing Exploratory Research (CSER) consortium, a component of NHGRI’s Genome Sequencing Program. CSER aims to investigate the actual use of genomic information in clinical care by defining conditions for which genome sequencing is clinically beneficial and by developing the necessary policies, procedures, and infrastructure to integrate genome‐sequence information into the clinical workflow. Nine CSER projects are using exome or genome sequencing for the care of patients with diverse conditions, including several types of cancer, cardiomyopathy, and developmental delay. A related program, the Return of Results Consortium [National Human Genome Research Institute e], is supporting seven studies examining the ethical, legal, and social issues involved in returning genomic research results to study participants and their families. Another new NHGRI genomic medicine project, conducted jointly with the National Institute of Child Health and Human Development, is building upon successful early CSER experiences to explore the implications and challenges of using genome sequencing in newborns [National Institute of Child Health and Human Development]. Four collaborating projects are acquiring and analyzing genome sequences that considerably expand the scale of data available for analysis in the newborn period; in addition, they are investigating the ethical, legal, and social implications of the possible implementation of genome sequencing as part of newborn medical care. The two remaining programs mentioned above arose directly from NHGRI’s consultations with the biomedical research community. The

6

AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS)

INVITED COMMENT

medicine implementation, diffusion, and sustainability.

2010. Rapid targeted mutational analysis of human tumours: a clinical platform to guide personalized cancer medicine. EMBO Mol Med 2:146–158. Eccles MP, Foy R, Sales A, Wensing M, Mittman B. 2012. Implementation Science six years on—Our evolving scope and common reasons for rejection without review. Implement Sci 7:71. Feero WG, Green ED. 2011. Genomics education for health care professionals in the 21st century. J Am Med Assoc 306:989–990. Gahl WA, Markello TC, Toro C, Fajardo KF, Sincan M, Gill F, Carlson‐Donohoe H, Gropman A, Pierson TM, Golas G, Wolfe L, Groden C, Godfrey R, Nehrebecky M, Wahl C, Landis DM, Yang S, Madeo A, Mullikin JC, Boerkoel CF, Tifft CJ, Adams D. 2012. The National Institutes of Health Undiagnosed Diseases Program: Insights into rare diseases. Genet Med 14:51–59. Green ED, Guyer MS, National Human Genome Research Institute. 2011. Charting a course for genomic medicine from base pairs to bedside. Nature 470:204–213. Lindhurst MJ, Sapp JC, Teer JK, Johnston JJ, Finn EM, Peters K, Turner J, Cannons JL, Bick D, Blakemore L, Blumhorst C, Brockmann K, Calder P, Cherman N, Deardorff MA, Everman DB, Golas G, Greenstein RM, Kato BM, Keppler‐Noreuil KM, Kuznetsov SA, Miyamoto RT, Newman K, Ng D, O’Brien K, Rothenberg S, Schwartzentruber DJ, Singhal V, Tirabosco R, Upton J, Wientroub S, Zackai EH, Hoag K, Whitewood‐Neal T, Robey PG, Schwartzberg PL, Darling TN, Tosi LL, Mullikin JC, Biesecker LG. 2011. A mosaic activating mutation in AKT1 associated with the Proteus syndrome. N Engl J Med 365:611– 619. Manolio TA, Murray MF, for the Inter‐Society Coordinating Committee on Practitioner Education in Genomics. 2014. The growing role of professional societies in educating clinicians in genomics. Genetics in Medicine (in press). Manolio TA, Chisholm RL, Ozenberger B, Roden DM, Williams MS, Wilson R, Bick D, Bottinger EP, Brilliant MH, Eng C, Frazer KA, Korf B, Ledbetter DH, Lupski JR, Marsh C, Mrazek D, Murray MF, O’Donnell PH, Rader DJ, Relling MV, Shuldiner AR, Valle D, Weinshilboum R, Green ED, Ginsburg GS. 2013. Implementing genomic medicine in the clinic: The future is here. Genet Med 15:258–267. McCarty CA, Chisholm RL, Chute CG, Kullo IJ, Jarvik GP, Larson EB, Li R, Masys DR, Ritchie MD, Roden DM, Struewing JP, Wolf WA, eMERGE Team. 2011. The eMERGE Network: a consortium of biorepositories linked to electronic medical records data for conducting genomic studies. BMC Med Genomics 4:13. Ng SB, Bigham AW, Buckingham KJ, Hannibal MC, McMillin MJ, Gildersleeve HI, Beck AE, Tabor HK, Cooper GM, Mefford HC, Lee C, Turner EH, Smith JD, Rieder MJ, Yoshiura K, Matsumoto N, Ohta T, Niikawa N, Nickerson DA, Bamshad MJ, Shendure J. 2010. Exome sequencing identifies MLL2 mutations as a cause of Kabuki syndrome. Nat Genet 42:790–793.

ClinGen project aims to fill an urgent need for a systematic approach to developing and disseminating consensus information about genomic variants relevant for clinical care, much as the CPIC program does for pharmacogenomics. To date, such efforts have mostly been pursued independently by individual groups, with investigators often evaluating the same assays, assessing the same evidence, and in most cases coming to the same conclusions, all in a highly duplicative fashion within and across sites. ClinGen was established to develop a consensus process for identifying genomic variants that are relevant for clinical care and to incorporate this information into an accessible electronic resource. The rationale for this project is described in another paper in this special issue [Ramos et al., 2014].

The ClinGen project aims to fill an urgent need for a systematic approach to developing and disseminating consensus information about genomic variants relevant for clinical care, much as the CPIC program does for pharmacogenomics. Lastly, the potential for “early adopter” genomic medicine sites to share their experiences and test the generalizability of their approaches in diverse clinical settings led NHGRI to solicit a series of genomic medicine demonstration projects [National Human Genome Research Institute f]. The recently funded IGNITE network is designed to expand and link existing genomic medicine efforts, develop new collaborative projects in diverse settings and populations, contribute to the evidence base supporting the use of genomic information for clinical care, and share best practices for genomic

SUMMARY The incorporation of genomic technologies and findings into routine clinical care holds considerable potential for personalizing medical treatments and enhancing the effectiveness of healthcare, but significant potential pitfalls abound. Chief among these is that the use of genomic information may not improve clinical outcomes, and almost certainly will not in every instance. Evidence supporting the utility of genomic information thus needs to be carefully assessed, while at the same time avoiding unreasonable expectations for exhaustive evidence for every genomic variant that may influence human health and disease. Potential misuses of genomic information that cause unnecessary worry, discrimination, increased medical costs, or diverted resources also need to be recognized and avoided. NHGRI expects the systematic set of research programs outlined above, in conjunction with additional future projects now in early design phases, will play key roles in addressing the major questions and barriers associated with genomic medicine implementation. In doing so, these efforts will provide a valuable complement to the highly successful basic research enterprise that has made such genomic advances conceivable.

REFERENCES Bainbridge MN, Wiszniewski W, Murdock DR, Friedman J, Gonzaga‐Jauregui C, Newsham I, Reid JG, Fink JK, Morgan MB, Gingras MC, Muzny DM, Hoang LD, Yousaf S, Lupski JR, Gibbs RA. 2011. Whole‐genome sequencing for optimized patient management. Sci Transl Med 3:87re3. Denny JC, Ritchie MD, Basford MA, Pulley JM, Bastarache L, Brown‐Gentry K, Wang D, Masys DR, Roden DM, Crawford DC. 2010. PheWAS: demonstrating the feasibility of a phenome‐wide scan to discover gene‐disease associations. Bioinformatics 26:1205– 1210. Dias‐Santagata D, Akhavanfard S, David SS, Vernovsky K, Kuhlmann G, Boisvert SL, Stubbs H, McDermott U, Settleman J, Kwak EL, Clark JW, Isakoff SJ, Sequist LV, Engelman JA, Lynch TJ, Haber DA, Louis DN, Ellisen LW, Borger DR, Iafrate AJ.

INVITED COMMENT

AMERICAN JOURNAL OF MEDICAL GENETICS PART C (SEMINARS IN MEDICAL GENETICS)

National Human Genome Research Institute (a). Genomic Medicine Activities. https://www. genome.gov/27549225 Accessed Oct. 13, 2013. National Human Genome Research Institute (b). Inter‐Society Coordinating Committee for Practitioner Education in Genomics. http:// www.genome.gov/27554614 Accessed Oct. 16, 2013. National Human Genome Research Institute (c). NHGRI Genome Sequencing Program. http://www.genome.gov/10001691 Accessed Oct. 16, 2013 National Human Genome Research Institute (d). RFA HG‐10‐009, The Electronic Medical Records and Genomics (eMERGE) Network, Phase II – Study Investigators. http://grants.nih.gov/grants/guide/rfa‐files/ RFA‐HG‐10‐009.html Accessed Oct. 16, 2013 National Human Genome Research Institute (e). RFA HG‐11‐003, Development of a Preliminary Evidence Base to Inform Decision‐making about Returning Research Results to Participants in Genomic Studies. http:// grants.nih.gov/grants/guide/rfa‐files/RFA‐HG‐11‐003.html Accessed Oct. 30, 2013. National Human Genome Research Institute, National Institute for Child Health and Human Development, Office of Rare Disease Research. Newborn Screening in the Genomic Era: Setting a Research Agenda. http://www.nichd.nih.gov/about/ meetings/2010/pages/121410.aspx Accessed Oct. 12, 2013.

National Institute of Child Health and Human Development and National Human Genome Research Institute. RFA HD‐13‐010, Genomic Sequencing and Newborn Screening Disorders. http://grants.nih.gov/grants/ guide/rfa‐files/RFA‐HD‐13‐010.html Accessed Oct. 30, 2013. National Institutes of Health (a). Office of Strategic Coordination—The Common Fund. Undiagnosed diseases. http://commonfund.nih. gov/diseases/ Accessed Oct. 30, 2013. National Human Genome Research Institute (f). Genomic Medicine Pilot Demonstration Projects. http://grants.nih.gov/grants/guide/ rfa‐files/RFA‐HG‐12‐006.html Accessed Jan. 29, 2014. National Institutes of Health (b). RFA RM‐13‐004, Clinical Sites for an Undiagnosed Diseases Network. http://grants.nih. gov/grants/guide/rfa‐files/RFA‐RM‐13‐004.html Accessed Nov. 23, 2013. Pharmacogenomics Research Network. Network‐ Wide Projects. http://pgrn.org/display/ pgrnwebsite/Network‐wideþProjects. Accessed Oct. 30, 2013. Ramos EM, Din‐Lovinescu C, Berg JS, Brooks LD, Duncanson A, Dunn M, Good P, Hubbard T, Jarvik GP, O’Donnell C, Sherry ST, Aronson N, Biesecker L, Blumberg B, Calonge1 N, Colhoun HM, Epstein RS, Flicek P, Gordon ES, Green ED, Green RC, Hurles M, Kawamoto K, Knaus W, Ledbetter DH, Levy HP, Lyon E, Maglott D, McLeod HL, Rahman N, Randhawa G, Wicklund C, Manolio TA, Chisholm RL,

7

Williams MS. 2014. Characterizing genetic variants for clinical action. Am J Med Genet Part C (in press). Relling MV, Klein TE. 2011. CPIC: Clinical pharmacogenetics implementation consortium of the pharmacogenomics research network. Clin Pharmacol Ther 89:464–467. Ritchie MD, Denny JC, Crawford DC, Ramirez AH, Weiner JB, Pulley JM, Basford MA, Brown‐Gentry K, Balser JR, Masys DR, Haines JL, Roden DM. 2010. Robust replication of genotype–phenotype associations across multiple diseases in an electronic medical record. Am J Hum Genet 86:560– 572. St. Hilaire C, Ziegler SG, Markello TC, Brusco A, Groden C, Gill F, Carlson‐Donohoe H, Lederman RJ, Chen MY, Yang D, Siegenthaler MP, Arduino C, Mancini C, Freudenthal B, Stanescu HC, Zdebik AA, Chaganti RK, Nussbaum RL, Kleta R, Gahl WA, Boehm M. 2011. NT5E mutations and arterial calcifications. N Engl J Med 364:432–442. Worthey EA, Mayer AN, Syverson GD, Helbling D, Bonacci BB, Decker B, Serpe JM, Dasu T, Tschannen MR, Veith RL, Basehore MJ, Broeckel U, Tomita‐Mitchell A, Arca MJ, Casper JT, Margolis DA, Bick DP, Hessner MJ, Routes JM, Verbsky JW, Jacob HJ, Dimmock DP. 2011. Making a definitive diagnosis: successful clinical application of whole exome sequencing in a child with intractable inflammatory bowel disease. Genet Med 13:255–262.

Leading the way to genomic medicine.

The National Human Genome Research Institute, in close collaboration with its research community, is pursuing an ambitious research agenda to facilita...
109KB Sizes 3 Downloads 3 Views