Comparing models of delivery for cancer genetics services among patients receiving primary care who meet criteria for genetic evaluation in two healthcare systems: BRIDGE randomized controlled trial

Kimberly A Kaphingst, Wendy Kohlmann, Rachelle Lorenz Chambers, Melody S Goodman, Richard Bradshaw, Priscilla A Chan, Daniel Chavez-Yenter, Sarah V Colonna, Whitney F Espinel, Jessica N Everett, Amanda Gammon, Eric R Goldberg, Javier Gonzalez, Kelsi J Hagerty, Rachel Hess, Kelsey Kehoe, Cecilia Kessler, Kadyn E Kimball, Shane Loomis, Tiffany R Martinez, Rachel Monahan, Joshua D Schiffman, Dani Temares, Katie Tobik, David W Wetter, Devin M Mann, Kensaku Kawamoto, Guilherme Del Fiol, Saundra S Buys, Ophira Ginsburg, BRIDGE research team, Kimberly A Kaphingst, Wendy Kohlmann, Rachelle Lorenz Chambers, Melody S Goodman, Richard Bradshaw, Priscilla A Chan, Daniel Chavez-Yenter, Sarah V Colonna, Whitney F Espinel, Jessica N Everett, Amanda Gammon, Eric R Goldberg, Javier Gonzalez, Kelsi J Hagerty, Rachel Hess, Kelsey Kehoe, Cecilia Kessler, Kadyn E Kimball, Shane Loomis, Tiffany R Martinez, Rachel Monahan, Joshua D Schiffman, Dani Temares, Katie Tobik, David W Wetter, Devin M Mann, Kensaku Kawamoto, Guilherme Del Fiol, Saundra S Buys, Ophira Ginsburg, BRIDGE research team

Abstract

Background: Advances in genetics and sequencing technologies are enabling the identification of more individuals with inherited cancer susceptibility who could benefit from tailored screening and prevention recommendations. While cancer family history information is used in primary care settings to identify unaffected patients who could benefit from a cancer genetics evaluation, this information is underutilized. System-level population health management strategies are needed to assist health care systems in identifying patients who may benefit from genetic services. In addition, because of the limited number of trained genetics specialists and increasing patient volume, the development of innovative and sustainable approaches to delivering cancer genetic services is essential.

Methods: We are conducting a randomized controlled trial, entitled Broadening the Reach, Impact, and Delivery of Genetic Services (BRIDGE), to address these needs. The trial is comparing uptake of genetic counseling, uptake of genetic testing, and patient adherence to management recommendations for automated, patient-directed versus enhanced standard of care cancer genetics services delivery models. An algorithm-based system that utilizes structured cancer family history data available in the electronic health record (EHR) is used to identify unaffected patients who receive primary care at the study sites and meet current guidelines for cancer genetic testing. We are enrolling eligible patients at two healthcare systems (University of Utah Health and New York University Langone Health) through outreach to a randomly selected sample of 2780 eligible patients in the two sites, with 1:1 randomization to the genetic services delivery arms within sites. Study outcomes are assessed through genetics clinic records, EHR, and two follow-up questionnaires at 4 weeks and 12 months after last genetic counseling contactpre-test genetic counseling.

Discussion: BRIDGE is being conducted in two healthcare systems with different clinical structures and patient populations. Innovative aspects of the trial include a randomized comparison of a chatbot-based genetic services delivery model to standard of care, as well as identification of at-risk individuals through a sustainable EHR-based system. The findings from the BRIDGE trial will advance the state of the science in identification of unaffected patients with inherited cancer susceptibility and delivery of genetic services to those patients.

Trial registration: BRIDGE is registered as NCT03985852 . The trial was registered on June 6, 2019 at clinicaltrials.gov .

Keywords: Genetic services; Health technology; Population health management; Primary care.

Conflict of interest statement

JDS is co-founder and shareholder in ItRunsInMyFamily.com, and co-founder and shareholder in PEEL Therapeutics, Inc.

KK2 reports honoraria, consulting, sponsored research, licensing, or co-development outside the submitted work in the past 3 years with McKesson InterQual, Hitachi, Pfizer, Premier, Klesis Healthcare, RTI International, Mayo Clinic, the University of Washington, the University of California at San Francisco, MD Aware, and the U.S. Office of the National Coordinator for Health IT (via ESAC and Security Risk Solutions) in the area of health information technology. KK2 was also an unpaid board member of the non-profit Health Level Seven International health IT standard development organization, he is an unpaid member of the U.S. Health Information Technology Advisory Committee, and he has helped develop a number of health IT tools which may be commercialized to enable wider impact. None of these relationships have direct relevance to the manuscript but are reported in the interest of full disclosure.

Figures

Fig. 1
Fig. 1
Study flow diagram for BRIDGE protocol

References

    1. King M-C, Marks JH, Mandell JB. The New York breast Cancer study group. Breast and ovarian cancer risks due to inherited mutations in BRCA1 and BRCA2. Science. 2003;302(5645):643–646. doi: 10.1126/science.1088759.
    1. Petrucelli N, Daly M, Pal T, et al. BRCA1- and BRCA2-associated hereditary breast and ovarian Cancer. In: Adam M, Ardinger H, Pagon R, et al., editors. GeneReviews. Seattle: University of Washington; 2016.
    1. Giardiello F, Allen J, Axilbund J, Boland C, Burke C, Burt R, Church JM, Dominitz JA, Johnson DA, Kaltenbach T, Levin TR, Lieberman DA, Robertson DJ, Syngal S, Rex DK, US Multi-Society Task Force on Colorectal Cancer Guidelines on genetic evaluation and management of lynch syndrome: a consensus statement by the US multi-society task force on colorectal Cancer. Gastroenterology. 2014;147(2):502–506. doi: 10.1053/j.gastro.2014.04.001.
    1. Curras-Freixes M, Inglada-Perez L, Mancikova V, Montero-Conde C, Leton R, et al. Recommendations for somatic and germline genetic testing of single pheochoromocytoma and paraganglioma based on findings from a series of 329 patients. J Med Genet. 2015;52(10):647–656. doi: 10.1136/jmedgenet-2015-103218.
    1. Brito J, Asi N, Bancos I, Gionfriddo M, Zeballos-Palacios C, et al. Testing for germline mutations in sporadic pheocromocytoma/paraganglioma: a systematic review. Clin Endocrinol. 2015;82(3):338–345. doi: 10.1111/cen.12530.
    1. Judkins T, Leclair B, Bowles K, Gutin N, Trost J, McCulloch J, Bhatnagar S, Murray A, Craft J, Wardell B, Bastian M, Mitchell J, Chen J, Tran T, Williams D, Potter J, Jammulapati S, Perry M, Morris B, Roa B, Timms K. Development and analytical validation of a 25-gene next generation sequencing panel that includes the BRCA1 and BRCA2 genes to assess hereditary cancer risk. BMC Cancer. 2015;15(1):215. doi: 10.1186/s12885-015-1224-y.
    1. National Cancer Institute. Genetics of Breast and Gynecologic Cancers Rockville: National Cancer Institute; 2015 [Available from: .
    1. Zhang S, Royer R, Li S, McLaughlin J, Rosen B, Risch H, et al. Frequencies of BRCA1 and BRCA2 mutations among 1,342 unselected patients with invasive ovarian cancer. Gynecol Oncol. 2011;121(2):353–357. doi: 10.1016/j.ygyno.2011.01.020.
    1. Hampel H, Frankel W, Martin E, et al. Screening for the lynch syndrome (hereditary nonpolyposis colorectal cancer) N Engl J Med. 2005;352(18):1851–1860. doi: 10.1056/NEJMoa043146.
    1. Yurgelun MB, Kulke MH, Fuchs CS, Allen BA, Uno H, Hornick JL, Ukaegbu CI, Brais LK, McNamara PG, Mayer RJ, Schrag D, Meyerhardt JA, Ng K, Kidd J, Singh N, Hartman AR, Wenstrup RJ, Syngal S. Cancer susceptibility gene mutations in individuals with colorectal cancer. J Clin Oncol. 2017;35(10):1086–1095. doi: 10.1200/JCO.2016.71.0012.
    1. Shindo K, Yu J, Suenaga M, Fesharakizadeh S, Cho C, Macgregor-Das A, Siddiqui A, Witmer PD, Tamura K, Song TJ, Navarro Almario JA, Brant A, Borges M, Ford M, Barkley T, He J, Weiss MJ, Wolfgang CL, Roberts NJ, Hruban RH, Klein AP, Goggins M. Deleterious germline mutations in patients with apparently sporadic pancreatic adenocarcinoma. J Clin Oncol. 2017;35(30):3382–3390. doi: 10.1200/JCO.2017.72.3502.
    1. Madelker D, et al. Mutation detection in patients with advanced cancer by universal sequencing of cancer-related genes in tumor and normal DNA vs guideline-based germline testing. J Am Med Assoc. 2017;318(9):825–835. doi: 10.1001/jama.2017.11137.
    1. Pritchard CC, Mateo J, Walsh MF, De Sarkar N, Abida W, Beltran H, et al. Inherited DNA-repair gene mutations in men with metastatic prostate cancer. N Engl J Med. 2016;375(5):443–453. doi: 10.1056/NEJMoa1603144.
    1. Hall MJ, Forman AD, Pilarski R, Wiesner G, Giri VN. Gene panel testing for inherited cancer risk. J Natl Compr Cancer Netw. 2014;12(9):1339–1346. doi: 10.6004/jnccn.2014.0128.
    1. Hiraki S, Rinella ES, Schnabel F, Oratz R, Ostrer H. Cancer risk assessment using genetic panel testing: considerations for clinical application. J Genet Couns. 2014;23(4):604–617. doi: 10.1007/s10897-014-9695-6.
    1. Kurian AW, Hare EE, Mills MA, Kingham KE, McPherson L, Whittemore AS, McGuire V, Ladabaum U, Kobayashi Y, Lincoln SE, Cargill M, Ford JM. Clinical evaluation of a multiple-gene sequencing panel for hereditary cancer risk assessment. J Clin Oncol. 2014;32(19):2001–2009. doi: 10.1200/JCO.2013.53.6607.
    1. Maxwell KN, Wubbenhorst B, D'Andrea K, et al. Prevalence of mutations in a panel of breast cancer susceptibility genes in BRCA1/2-negative patients with early-onset breast cancer. Genet Med. 2015;17:630–8.
    1. Desmond A, Kurian AW, Gabree M, Mills MA, Anderson MJ, Kobayashi Y, Horick N, Yang S, Shannon KM, Tung N, Ford JM, Lincoln SE, Ellisen LW. Clinical actionability of multigene panel testing for hereditary breast and ovarian cancer risk assessment. JAMA Oncol. 2015;1(7):943–951. doi: 10.1001/jamaoncol.2015.2690.
    1. Slavin TP, Niell-Swiller M, Solomon I, Nehoray B, Rybak C, Blazer KR, et al. Clinical application of multigene panels: challenges of next-generation counseling and cancer risk management. Front Oncol. 2015;5:208.
    1. Pennington KP, Swisher EM. Hereditary ovarian cancer: beyond the usual suspects. Gynecol Oncol. 2012;124(2):347–353. doi: 10.1016/j.ygyno.2011.12.415.
    1. Domchek SM, Bradbury A, Garber JE, Offit K, Robson ME. Multiplex genetic testing for cancer susceptibility: out on the high wire without a net? J Clin Oncol. 2013;31(10):1267–1270. doi: 10.1200/JCO.2012.46.9403.
    1. Mandelker D, Zhang L, Kemel Y, Stadler ZK, Joseph V, Zehir A, Pradhan N, Arnold A, Walsh MF, Li Y, Balakrishnan AR, Syed A, Prasad M, Nafa K, Carlo MI, Cadoo KA, Sheehan M, Fleischut MH, Salo-Mullen E, Trottier M, Lipkin SM, Lincoln A, Mukherjee S, Ravichandran V, Cambria R, Galle J, Abida W, Arcila ME, Benayed R, Shah R, Yu K, Bajorin DF, Coleman JA, Leach SD, Lowery MA, Garcia-Aguilar J, Kantoff PW, Sawyers CL, Dickler MN, Saltz L, Motzer RJ, O’Reilly EM, Scher HI, Baselga J, Klimstra DS, Solit DB, Hyman DM, Berger MF, Ladanyi M, Robson ME, Offit K. Mutation detection in patients with advanced Cancer by universal sequencing of Cancer-related genes in tumor and Normal DNA vs guideline-based Germline testing. JAMA. 2017;318(9):825–835. doi: 10.1001/jama.2017.11137.
    1. Pearlman R, Frankel W, Swanson B, et al. Prevalence and spectrum of germline cancer susceptibility gene mutations among patietns with early onset colorectal cancer. JAMA Oncol. 2017;3(4):464–471. doi: 10.1001/jamaoncol.2016.5194.
    1. Stoffel E, Koeppe E, Everett J, et al. Germline genetic features of young individuals with colorectal cancer. Gastroenterology. 2018;154(4):897–905. doi: 10.1053/j.gastro.2017.11.004.
    1. Kapoor N, Curcio L, Blakemore C, et al. Multigene panel testing detects equal rates of pathogenic BRCA1/2 mutations and has a higher diagnostic yield compared to limited BRCA1/2 analysis along in patients at risk for hereditary breast cancer. Ann Surg Oncol. 2015;22(10):3282–3288. doi: 10.1245/s10434-015-4754-2.
    1. Ricker C, Culver JO, Lowstuter K, Sturgeon D, Sturgeon JD, Chanock CR, Gauderman WJ, McDonnell KJ, Idos GE, Gruber SB. Increased yield of actionable mutations using multi-gene panels to assess hereditary cancer susceptibility in an ethnically diverse clinical cohort. Cancer Genet. 2016;209(4):130–137. doi: 10.1016/j.cancergen.2015.12.013.
    1. Susswein LR, Marshall ML, Nusbaum R, Vogel Postula KJ, Weissman SM, Yackowski L, Vaccari EM, Bissonnette J, Booker JK, Cremona ML, Gibellini F, Murphy PD, Pineda-Alvarez DE, Pollevick GD, Xu Z, Richard G, Bale S, Klein RT, Hruska KS, Chung WK. Pathogenic and likely pathogenic variant prevalence among the first 10,000 patients referred for next-generation cancer panel testing. Genet Med. 2016;18(8):823–832. doi: 10.1038/gim.2015.166.
    1. Yurgelun MB, Allen B, Kaldate RR, Bowles KR, Judkins T, Kaushik P, Roa BB, Wenstrup RJ, Hartman AR, Syngal S. Identification of a variety of mutations in Cancer predisposition genes in patients with suspected lynch syndrome. Gastroenterology. 2015;149(3):604–613. doi: 10.1053/j.gastro.2015.05.006.
    1. Giri VN, Knudsen K, Kelly W, et al. Role of genetic testing for inherited prostate cancer risk: Philadelphia prostate Cancer consensus conference 2017. J Clin Oncol. 2018;36(4):414–424. doi: 10.1200/JCO.2017.74.1173.
    1. Hampel H. Genetic counseling and cascade genetic testing in lynch syndrome. Familial Cancer. 2016;15(3):423–427. doi: 10.1007/s10689-016-9893-5.
    1. Samini G, Bernardini M, Brody L, Caga-Anan C, Campbell I, Chenevix-Trench G. Traceback: a proposed framework to increase identification and genetic counseling of BRCA1 and BRCA2 mutation carriers through family-based outreach. J Clin Oncol. 2017;35(20):2329–2337. doi: 10.1200/JCO.2016.70.3439.
    1. Katapodi M, Viassolo V, Caiata-Zufferey M, Nikolaidis C, Buhrer-Landolt R, Buerki N, et al. Cancer predisposition cascade screening for hereditary breast/ovarian cancer and lynch syndromes in Switzerland: study protocol. JMIR Res Prot. 2017;6(9):e184. doi: 10.2196/resprot.8138.
    1. Dinh T, Rosner B, Atwood J, Boland C, et al. Health benefits and cost-effectiveness of primary genetic screening for lynch syndrome in the general population. Cancer Prev Res. 2011;4(1):9–22. doi: 10.1158/1940-6207.CAPR-10-0262.
    1. Li Y, Arellano A, Bare L, et al. A multigene test could cost-effectively help extend life expectancy for women at risk of hereditary breast cancer. Value Health. 2017;20(4):547–555. doi: 10.1016/j.jval.2017.01.006.
    1. Luba D, Disario J, Rock C, et al. Community practice implementation of a self-administered version of PREMM1,2,6 to assess risk for lynch syndrome. Clin Gastroenterol Hepatol. 2018;16(1):49–58. doi: 10.1016/j.cgh.2017.06.038.
    1. Gou F, et al. Use of BRCA mutation test in the U.S. 2004-2014. Prev Med. 2017;52(6):702–9.
    1. Cropper C, Woodson AH, Arun B, Barcenas C, Litton J, Noblin S, Liu D, Park M, Daniels M. Evaluating the NCCN criteria for recommending BRCA1 and BRCA2 genetic testing in patients with breast cancer. J Natl Compr Cancer Netw. 2017;15(6):797–803. doi: 10.6004/jnccn.2017.0107.
    1. Gilpin C, Carson N, Hunter A. A preliminary validation of a family history assessment form to select women at risk for breast or ovarian cancer for referral to a genetics center. Clin Genet. 2000;58(4):299–308. doi: 10.1034/j.1399-0004.2000.580408.x.
    1. Parmigiani G, Chen S, Iversen E., Jr Validity of models for predicting BRCA1 and BRCA2 mutations. Ann Intern Med. 2007;147(7):441–450. doi: 10.7326/0003-4819-147-7-200710020-00002.
    1. Bellcross C, Lemke A, Paper L, et al. Evaluation of a breast/ovarian cancer genetics referral screening tool in a mammography population. Genet Med. 2009;11(11):783–789. doi: 10.1097/GIM.0b013e3181b9b04a.
    1. Hoskins K, Zwaagstra A, Ranz M. Validation of a tool for identifying women at high risk for hereditary breast cancer in population-based screening. Cancer. 2006;107(8):1769–1776. doi: 10.1002/cncr.22202.
    1. Ashton-Prolla P, Giacomazzi J, Schmidt A, et al. Development and validation of a simple questionnaire for the identification of hereditary breast cancer in primary care. BMC Cancer. 2009;9(1):283. doi: 10.1186/1471-2407-9-283.
    1. Khoury MJ, Feero WG, Valdez R. Family history and personal genomics as tools for improving health in an era of evidence-based medicine. Am J Prev Med. 2010;39(2):184–188. doi: 10.1016/j.amepre.2010.03.019.
    1. U.S. Preventive Services Task Force Risk assessment, genetic counseling, and genetic testing for BRCA-related cancer: recommendation statement. Am Fam Physician. 2020;101(4):233–238.
    1. Centers for Medicare & Medicaid Services. Affordable Care Act Implementation FAQs - Set 12 Baltimore: Centers for Medicare & Medicaid Services. . Accessed 1 Mar 2021.
    1. Acheson LS, Weisner GL, Zyzanski SJ, Goodwin MA, Stange KC. Family history-taking in community family practice: implications for genetic screening. Genet Medicine. 2000;2(3):180–185. doi: 10.1097/00125817-200005000-00004.
    1. Fuller M, Myers M, Webb T, Tabangin M, Prows C. Primary care providers' responses to patient-centered family history. J Genet Couns. 2010;19(1):84–96. doi: 10.1007/s10897-009-9264-6.
    1. Valdez R, Yoon PW, Qureshi N, Green RF, Khoury MJ. Family history in public health practice: a genomic tool for disease prevention and health promotion. Annu Rev Public Health. 2010;31(1):69–87. doi: 10.1146/annurev.publhealth.012809.103621.
    1. Ginsburg O, Ashton-Prolla P, Cantor A, Mariosa D, Brennan P. The role of genomics in global cancer prevention. Nat Rev Clin Oncol. 2020; ePub before press.
    1. Grindedal E, Herabm C, Karsrud I, et al. Current guidelines for BRCA testing of breast cancer patietns are insufficient to detect all mutation carriers. BMC Cancer. 2017;17(1):438. doi: 10.1186/s12885-017-3422-2.
    1. Hull L, Haas J, Simon S, et al. Provider discussions of genetic tests with U.S. womend at risk for BRCA mutation. Am J Prev Med. 2018;54(2):221–228. doi: 10.1016/j.amepre.2017.10.015.
    1. Acheson L. Fostering applications of genetics in primary care: what will it take? Genet Med. 2003;5(2):63–65. doi: 10.1097/01.GIM.0000056946.67707.67.
    1. Rich EC, Burke W, Heaton CJ, Haga S, Pinsky L, Short P, et al. Reconsidering the family history in primary care. J Gen Intern Med. 2004;19(3):273–280. doi: 10.1111/j.1525-1497.2004.30401.x.
    1. Green RF, Olney RS. Connecting generations: family history an important tool in pediatrics, public health. AAP News. 2007;28:26.
    1. Kelly KM, Ferketich AK, Sturm AC, Porter K, Sweet K, Kemp K, Schwirian P, Westman JA. Cancer risk and risk communication in urban, lower-income neighborhoods. Prev Med. 2009;48(4):392–396. doi: 10.1016/j.ypmed.2009.01.009.
    1. Kelly KM, Love MM, Pearce KA, Porter K, Barron MA, Andrykowski M. Cancer risk assessment by rural and Appalachian family medicine physicians. J Rural Health. 2009;25(4):372–377. doi: 10.1111/j.1748-0361.2009.00246.x.
    1. Delikurt T, Williamson G, Anastasiadou V, Skirton H. A systematic review of factors that act as barriers to patient referral to genetic services. Eur J Hum Genet. 2015;23(6):739–745. doi: 10.1038/ejhg.2014.180.
    1. Mikat-Stevens NA, Larson IA, Tarini BA. Primary-care providers' perceived barriers to integration of genetics services: a systematic review of the literature. Genet Med. 2015;17(3):169–176. doi: 10.1038/gim.2014.101.
    1. Mowery DL, Kawamoto K, Bradshaw R, Kohlmann W, Schiffman JD, Weir C, Borbolla D, Chapman WW, del Fiol G. Determining onset for familial breast and colorectal cancer from family history comments in the electronic health record. AMIA Jt Summits Transl Sci Proc. 2019;2019:173–181.
    1. Gupta S, Provenzale D, Llor X, Halverson AL, Grady W, Chung DC, Haraldsdottir S, Markowitz AJ, Slavin Jr TP, Hampel H, Ness RM, Weiss JM, Ahnen DJ, Chen LM, Cooper G, Early DS, Giardiello FM, Hall MJ, Hamilton SR, Kanth P, Klapman JB, Lazenby AJ, Lynch PM, Mayer RJ, Mikkelson J, Peter S, Regenbogen SE, Dwyer MA, Ogba N. Genetic/familial high-risk assessment: colorectal, version 2.2019: featured updates to the NCCN guidelines. J Natl Compr Cancer Netw. 2019;17(9):1032–1041. doi: 10.6004/jnccn.2019.0044.
    1. Daly MB, Pilarski R, Yurgelun MB, Berry MP, Buys SS, Dickson P, Domchek SM, Elkhanany A, Friedman S, Garber JE, Goggins M, Hutton ML, Khan S, Klein C, Kohlmann W, Kurian AW, Laronga C, Litton JK, Mak JS, Menendez CS, Merajver SD, Norquist BS, Offit K, Pal T, Pederson HJ, Reiser G, Shannon KM, Visvanathan K, Weitzel JN, Wick MJ, Wisinski KB, Dwyer MA, Darlow SD. Genetic/familial high-risk assessment: breast, ovarian, and pancreatic, version 1.2020: featured updates to the NCCN guidelines. J Natl Compr Cancer Netw. 2020;18(4):380–391. doi: 10.6004/jnccn.2020.0017.
    1. Del Fiol G, Kohlmann W, Bradshaw R, Weir C, Flynn M, Hess R, et al. Standards-based clinical decision support platform to manage patients who meet guideline-based criteria for genetic evaluation of familial cancer. J Clin Oncol Clin Cancer Inform. 2020;4:1–9.
    1. Hoskovec JM, Bennett RL, Carey ME, DaVanzo JE, Dougherty MJ, Hahn SE, et al. Projecting the supply and demand for certified genetic counselors: a workforce study. J Genet Couns. 2018;27(1):16–20. doi: 10.1007/s10897-017-0158-8.
    1. Attard C, Carmany E, Trepanier A. Genetic counselor workflow study: the times are they a-changin? J Genet Couns. 2019;28(1):130–140. doi: 10.1002/jgc4.1041.
    1. Maiese D, Keehn A, Lyon M, Flannery D, Watson M. Current conditions in medical genetics practice. Genet Med. 2019;21(8):1874–1877. doi: 10.1038/s41436-018-0417-6.
    1. Buchanan A, Rahm A, Williams J. Alternate service delivery models in cancer genetic counseling: a mini-review. Front Oncol. 2016;6:120. doi: 10.3389/fonc.2016.00120.
    1. Kinney AY, Steffen LE, Brumbach BH, Kohlmann W, Du R, Lee JH, et al. Randomized noninferiority trial of telephone delivery of BRCA1/2 genetic counseling compared with in-person counseling: 1-year follow-up. J Clin Oncol. 2016;34(24):2914–24. doi: 10.1200/JCO.2015.65.9557.
    1. Schwartz MD, Valdimarsdottir HB, Peshkin BN, Mandelblatt JS, Nusbaum R, Huang A-T, Chang Y, Graves K, Isaacs C, Wood M, McKinnon W, Garber J, McCormick S, Kinney AY, Luta G, Kelleher S, Leventhal KG, Vegella P, Tong A, King L. Randomized noninferiority trial of telephone versus in-person genetic counseling for hereditary breast and ovarian cancer. J Clin Oncol. 2014;32(7):618–626. doi: 10.1200/JCO.2013.51.3226.
    1. Sutphen R, Davila B, Shappell H, Holtje T, Vadaparampil S, Friedman S, Toscano M, Armstrong J. Real world experience with cancer genetic counseling via telephone. Familial Cancer. 2010;9(4):681–689. doi: 10.1007/s10689-010-9369-y.
    1. McCuaig J, Armel S, Care M, Volenik A, Kim R, Metcalfe K. Next-generation service delivery: a scoping review of patient outcomes associated with alternative models of genetic counseling and genetic testing for hereditary cancer. Cancers. 2018;10(11):435. doi: 10.3390/cancers10110435.
    1. Voils C, Venne V, Weidenbacher H, Sperber N, Datta S. Comparison of telephone and televideo modes for delivery of genetic counseling: a randomized trial. J Genet Couns. 2018;27(2):339–348. doi: 10.1007/s10897-017-0189-1.
    1. Butrick M, Kelly S, Peshkin BN, Luta G, Nusbaum R, Hooker GW, Graves K, Feeley L, Isaacs C, Valdimarsdottir HB, Jandorf L, DeMarco T, Wood M, McKinnon W, Garber J, McCormick SR, Schwartz MD. Disparities in uptake of BRCA1/2 genetic testing in a randomized trial of telephone counseling. Genet Med. 2015;17(6):467–475. doi: 10.1038/gim.2014.125.
    1. Steffen L, Du R, Gammon A, Mandelblatt J, Kohlmann W, Lee J, et al. Genetic testing in a population-based sample of breast and ovarian cancer survivors from the REACH randomized trial: cost barriers and moderators of counseling mode. Cancer Epidemiol Biomark Prev. 2017;26(12):1772–1780. doi: 10.1158/1055-9965.EPI-17-0389.
    1. Cohen S, Huziak R, Gustafson S, Grubs R. Analysis of advantages, limitations, and barriers of genetic counseling service delivery models. J Genet Couns. 2016;25(5):1010–1018. doi: 10.1007/s10897-016-9932-2.
    1. Green MJ, Peterson SK, Baker MW, Friedman LC, Harper GR, Rubinstein WS, et al. Use of an educational computer program before genetic counseling for breast cancer susceptibility: Effects on duration and content of counseling sessions. Genet Med. 2005;7(4):221–229. doi: 10.1097/01.gim.0000159905.13125.86.
    1. Green MJ, Peterson SK, Baker MW, Harper GR, Friedman LC, Rubinstein WS, Mauger DT. Effect of a computer-based decision aid on knowledge, perceptions, and intentions about genetic testing for breast cancer susceptibility: a randomized controlled trial. JAMA. 2004;292(4):442–452. doi: 10.1001/jama.292.4.442.
    1. Trepanier A, Allain D. Models of service delivery for cancer genetic risk assessment and counseling. J Genet Couns. 2014;23(2):239–253. doi: 10.1007/s10897-013-9655-6.
    1. Keshavan M. Key players in the direct-to-consumer lab testing market: MedCityNews. 2016.
    1. van der Wouden C, Carere D. Maitland-van der zee a. consumer perceptions of interactions with primary care providers after direct-to-consumer personal genomic testing. Ann Intern Med. 2016;164(8):513–522. doi: 10.7326/M15-0995.
    1. Burke W, Trinidad S. The deceptive appeal of direct-to-consumer genetics. Ann Intern Med. 2016;164(8):564–565. doi: 10.7326/M16-0257.
    1. Tandy-Connor S, Guiltinan J, Kremply K, LaDuca H, Reineke P, Gutierrez S, et al. False-positive results released by direct-to-consumer genetic tests highlight the importance of clinical confirmation testing for appropriate patient care. Genet Med. 2018;20(12):1515–1521. doi: 10.1038/gim.2018.38.
    1. Hamilton J, Abdiwahab E, Edwards H, Fan M, Jdayani A, Breslau E. Primary care providers' cancer genetic-testing related knowledge, attitudes, and communication behaviors: a systematic review and research agenda. J Gen Intern Med. 2017;32(3):315–324. doi: 10.1007/s11606-016-3943-4.
    1. Doak CC, Doak LG, Root JH. Teaching patients with low literacy skills. 2. Philadelphia: J.B. Lippincott Company; 1996. Teaching Patients with Low Literacy Skills.
    1. Mayer RE, Dow GT, Mayer S. Multimedia learning in an interactive self-explaining environment: what works in the design of agent-based microworlds. J Educ Psychol. 2003;95(4):806–813. doi: 10.1037/0022-0663.95.4.806.
    1. Agency for Healthcare Research and Quality . Health literacy universal precautions toolkit. 2010.
    1. Bibault J-E, Chaix B, Nectoux P, Pienkowski A, Guillemase A, Brouard B. Healthcare ex Machina: are conversational agents ready for prime time in oncology? Clin Transl Rad Oncol. 2019;16:55–59. doi: 10.1016/j.ctro.2019.04.002.
    1. Fitzpatrick K, Darcy A, Vierhile M. Delivering cognitive behavior therapy to young adults with symptoms of depression and anxiety using a fully automated conversational agent (Woebot): a randomized controlled trial. JMIR Mental Health. 2017;4(2):e19. doi: 10.2196/mental.7785.
    1. Ly K, Ly A-M, Andersson G. A fully automated conversational agent for promoting mental wellbeing: a pilot RCT using mixed methods. Internet Interv. 2017;10:39–46. doi: 10.1016/j.invent.2017.10.002.
    1. Owens O, Felder T, Tavakoli A, Revels A, Friedman D, Hughes-Halbert C, Hébert JR. Evaluation of a computer-based decision aid for promoting informed prostate cancer screening decisions among African American men: iDecide. Am J Health Promot. 2018;33(2):267–278. doi: 10.1177/0890117118786866.
    1. Pereira J, Diaz O. Using health chatbots for behavior change: a mapping study. J Med Syst. 2019;43(5):135. doi: 10.1007/s10916-019-1237-1.
    1. Gordon E, Babu D, Laney D. The future is now: technology’s impact on the practice of genetic counseling. Am J Med Genet. 2018;178(1):15–23. doi: 10.1002/ajmg.c.31599.
    1. Biesecker B. Genetic counseling and the central tenets of practice. Cold Spring Harbor Perspect Med. 2020;10(3):a038968.
    1. Flannery D. Challenges and opportunities for effective delivery of clinical genetic services in the U.S. healthcare system. Curr Opin Pediatr. 2018;30(6):740–745. doi: 10.1097/MOP.0000000000000693.
    1. Rashkin M, Bowes J, Dunaway K, Dhaliwal J, Loomis E, Riffle S, et al. Genetic counseling, 2030: an on-demand service tailored to the needs of a price conscious, genetically literate, and busy world. J Genet Couns. 2019;28(2):456–465. doi: 10.1002/jgc4.1123.
    1. Schmidlen T, Schwartz M, Diloreto K, Kirchner H, Sturm A. Patient assessment of chatbots for the scalable delivery of genetic counseling. J Genet Couns. 2019;28(6):1166–1177. doi: 10.1002/jgc4.1169.
    1. Nadarzynski T, MIles O, Cowie A, Ridge D. Acceptability of artificial intelligence (AI)-led chatbot services in healthcare: a mixed methods study. Digital Health. 2019;5:1–12.
    1. Roberts MC, Kennedy AE, Chambers DA, Khoury MJ. The current state of implementation science in genomic medicine: opportunities for improvement. Genet Med. 2017;19(8):858–863. doi: 10.1038/gim.2016.210.
    1. Institute of Medicine . Unequal treatment: understanding racial and ethnic disparities in health Care. Washington, DC: National Academy Press; 2002.
    1. Halbert C, Harrison B. Genetic counseling among minority populations in the era of precision medicine. Am J Med Genet. 2018;178(1):68–74. doi: 10.1002/ajmg.c.31604.
    1. Pierle J, Mahon S. Genetic service delivery models: exploring approaches to care for families with hereditary cancer risk. Clin J Oncol Nurs. 2019;23(1):60–67. doi: 10.1188/19.CJON.60-67.
    1. Hall M, Olopade OI. Confronting genetic testing disparities: knowledge is power. J Am Med Assoc. 2005;293(14):1783–1785. doi: 10.1001/jama.293.14.1783.
    1. Hall MJ, Olopade OI. Disparities in genetic testing: thinking outside the BRCA box. J Clin Oncol. 2006;24(14):2197–2203. doi: 10.1200/JCO.2006.05.5889.
    1. Fisher E, Pratt R, Esch R, Kocher M, Wilson KR, Lee W, et al. The role of race and ethnicity in views toward and participation in genetic studies and precision medicine research in the United States: a systematic review of qualitative and quantitative studies. Mol Genet Gen Med. 2019;8(2):e1099.
    1. McCarthy AM, Bristol M, Domchek SM, Groeneveld PW, Kim Y, Motanya UN, et al. Health care segregation, physician recommendation, and racial disparities in BRCA1/2 testing among women with breast cancer. J Clin Oncol. 2016;34(22):2610–2618. doi: 10.1200/JCO.2015.66.0019.
    1. Alford SH, McBride CM, Reid RJ, Larson EB, Baxevanis AD, Brody LC. Participation in genetic testing research varies by social group. Public Health Gen. 2011;14(2):85–93. doi: 10.1159/000294277.
    1. Defining rural population. Available from: . Accessed 26 Feb 2021.
    1. Caldwell JT, Ford CL, Wallace SP, Wang MC, Takahashi LM. Intersection of living in a rural versus urban area and race/ethnicity in explaining access to health care in the United States. Am J Public Health. 2016;106(8):1463–1469. doi: 10.2105/AJPH.2016.303212.
    1. Bhuyan SS, Wang Y, Opoku S, Lin G. Rural-urban differences in acute myocardial infarction mortality: evidence from Nebraska. J Cardiovasc Dis Res. 2013;4(4):209–213. doi: 10.1016/j.jcdr.2014.01.006.
    1. Weaver KE, Geiger AM, Lu L, Case LD. Rural-urban disparities in health status among US cancer survivors. Cancer. 2013;119(5):1050–1057. doi: 10.1002/cncr.27840.
    1. Blake K, Moss J, Gaysynsky A, Srinivasan S, Croyle R. Making the case for investment in rural cancer control: an analysis of rural cancer incidence, mortality, and funding trends. Cancer Epidemiol Biomark Prev. 2017;26(7):992–997. doi: 10.1158/1055-9965.EPI-17-0092.
    1. Henley S, Anderson R, Thomas C, Massetti G, Peaker B, Richardson L. Invasive cancer incidence, 2004-2013, and deaths, 2006-2015, in nonmetropolitan and metropolitan counties - United States. MMWR. 2017;66(14):1–13. doi: 10.15585/mmwr.ss6614a1.
    1. Zahnd W, James A, Jenkins W, Izadi S, Fogleman A, Steward D, et al. Rural-urban differences in cancer incidence and trends in the United States. Cancer Epidemiol Biomark Prev. 2018;27(11):1265–1274. doi: 10.1158/1055-9965.EPI-17-0430.
    1. George R, Kovak K, Cox SL. Aligning policy to promote cascade genetic screening for prevention and early diagnosis of heritable diseases. J Genet Couns. 2015;24(3):388–399. doi: 10.1007/s10897-014-9805-5.
    1. Villegas C, Haga S. Access to genetic counselors in the southern United States. J Personalized Med. 2019;9(3):33. doi: 10.3390/jpm9030033.
    1. Radford C, Prince A, Lewis K, Pal T. Factors which impact the delivery of genetic risk assessment services focused on inherited cancer genomics: expanding the role and reach of certified genetics professionals. J Genet Couns. 2014;23(4):522–530. doi: 10.1007/s10897-013-9668-1.
    1. Glasgow R, Klesges L, Dzewaltowski D, Estabrooks P, TM V. Evaluating the impact of health promotion programs: using the RE-AIM framework to form summary measures for decision making involving complex issues. Health Educ Res. 2006;21(5):688–694. doi: 10.1093/her/cyl081.
    1. Glasgow RE, Vogt TM, Boles SM. Evaluating the public health impact of health promotion interventions: the RE-AIM framework. Am J Public Health. 1999;89(9):1322–1327. doi: 10.2105/AJPH.89.9.1322.
    1. National Comprehensive Cancer Network . Breast Cancer Screening and Diagnosis Version I.2017. 2017.
    1. National Comprehensive Cancer Network . Genetic/Familial High-Risk Assessment: Colorectal Version 3.2017. 2017.
    1. National Comprehensive Cancer Network . Genetic/familial high-risk assessment: breast/ovarian, Version I 2018. 2018.
    1. National Comprehensive Cancer Network . Colorectal Cancer Screening Version 2.2017. 2017.
    1. National Comprehensive Cancer Network . Prostate Cancer Early Detection Version 2.2017. 2017.
    1. DeMarco T, Peshkin B, Mars B, Tercyak K. Patient satisfaction with cancer genetic counseling: a psychometric anaysis of the genetic counseling satisfaction scale. J Genet Couns. 2004;13(4):293–304. doi: 10.1023/B:JOGC.0000035523.96133.bc.
    1. Peshkin BN, Kelly S, Nusbaum RH, Similuk M, DeMarco TA, Hooker GW, et al. Patient perceptions of telephone vs. in-person BRCA1/BRCA2 genetic counseling. J Genet Couns. 2015;25:472–482. doi: 10.1007/s10897-015-9897-6.
    1. Kaphingst KA, McBride CM, Wade CH, Baxevanis AD, Reid RJ, Larson EB, et al. Patients' understanding of and responses to multiplex genetic susceptibility test results. Genet Med. 2012;14(7):681–687. doi: 10.1038/gim.2012.22.
    1. Lumish HS, Steinfeld H, Koval C, Russo D, Levinson E, Wynn J, Duong J, Chung WK. Impact of panel gene testing for hereditary breast and ovarian cancer on patients. J Genet Couns. 2017;26(5):1116–1129. doi: 10.1007/s10897-017-0090-y.
    1. Roberts JS, Gornick MC, Carere DA, Uhlmann WR, Ruffin MT, Green RC. Direct-to-consumer genetic testing: user motivations, decision making, and perceived utility of results. Public Health Genomics. 2017;20(1):36–45. doi: 10.1159/000455006.
    1. Brehaut J, O'Connor A, Wood T, Hack T, Siminoff L, Gordon E, et al. Validation of a decision regret scale. Med Decis Mak. 2003;23(4):281–292. doi: 10.1177/0272989X03256005.
    1. Underhill-Blazey M, Stopfer J, Chittenden A, Nayak MM, Lansang K, Lederman R, Garber J, Gundersen DA. Development and testing of the KnowGene scale to assess general genetic knowledge related to multigene panel testing. Patient Educ Couns. 2019;102(8):1558–1564. doi: 10.1016/j.pec.2019.04.014.
    1. Cella D, Hughes C, Peterman A, Chang C-H, Peshkin BN, Schwartz MD, Wenzel L, Lemke A, Marcus AC, Lerman C. A brief assessment of concerns associated with genetic testing for cancer: the multidimensional impact of Cancer risk assessment (MICRA) questionnaire. Health Psychol. 2002;21(6):564–572. doi: 10.1037/0278-6133.21.6.564.
    1. Fagerlin A, Zikmund-Fisher BJ, Ubel PA, Jankovic A, Derry HA, Smith DM. Measuring numeracy without a math test: development of the subjective numeracy scale. Med Decis Mak. 2007;27(5):672–680. doi: 10.1177/0272989X07304449.
    1. Chew LD, Griffin JM, Partin MR, Noorbaloochi S, Grill JP, Snyder A, Bradley KA, Nugent SM, Baines AD, VanRyn M. Validation of screening questions for limited health literacy in a large VA outpatient population. J Gen Intern Med. 2008;23(5):561–566. doi: 10.1007/s11606-008-0520-5.
    1. Norman CD, Skinner HA. eHEALS: the eHealth literacy scale. J Med Internet Res. 2006;8(4):e27. doi: 10.2196/jmir.8.4.e27.
    1. Parrott R, Silk K, Krieger JR, Harris T, Condit C. Behavioral health outcomes associated with religious faith and media exposure about human genetics. Health Commun. 2004;16(1):29–45. doi: 10.1207/S15327027HC1601_3.
    1. Hesse BW, Nelson DE, Kreps GL, Croyle RT, Arora NK, Rimer BK, Viswanath K. Trust and sources of health information: the impact of the internet and its implications for health care providers: findings from the first health information National Trends Survey. Arch Intern Med. 2005;165(22):2618–2624. doi: 10.1001/archinte.165.22.2618.
    1. Nelson DE, Kreps GL, Hesse BW, Croyle RT, Willis G, Arora NK, Rimer B, Vish Viswanath K, Weinstein N, Alden S. The health information National Trends Survey (HINTS): development, design, and dissemination. J Health Commun. 2004;9(5):443–460. doi: 10.1080/10810730490504233.
    1. Dutta-Bergman M. Trusted online sources of health information: differences in demographics, health beliefs, and health-information orientation. J Med Internet Res. 2003;5(3):e21. doi: 10.2196/jmir.5.3.e21.
    1. Conway LG, Woodard SR, Zubrod A. Social psychological measurements of COVID-19: coronavirus perceived threat, government response, impacts, and experiences questionnaires. 2020.
    1. National Cancer Institute . Cancer Moonshot Blue Ribbon Panel Report 2016. Bethesda: National institutes of health, National Cancer Institute; 2016.

Source: PubMed

3
Abonneren