1
|
Li H, Zhao Y, Li W, Wang W, Zhi S, Wu Y, Zhong Q, Wang R, Sun J. A WeChat-Based Decision Aid Intervention to Promote Informed Decision-Making for Family Members Regarding the Genetic Testing of Patients With Colorectal Cancer: Randomized Controlled Trial. J Med Internet Res 2025; 27:e60681. [PMID: 40258273 PMCID: PMC12053134 DOI: 10.2196/60681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 10/22/2024] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Identifying patients with inherited colorectal cancer (CRC) syndromes offers many potential benefits. However, individuals often experience decisional conflict regarding genetic testing for CRC, and the uptake rate remains low. Given the growing popularity of genetic testing and the increasing demands on genetic service providers, strategies are needed to promote informed decision-making, increase genetic testing uptake among at-risk individuals, and ensure the rational use of genetic service resources. OBJECTIVE This study aims to determine whether a decision aid (DA) tool could promote informed decision-making among family members regarding the genetic testing of a patient with CRC. METHODS A single-center, parallel-group, randomized controlled trial was conducted. We randomized 82 family members of patients with CRC, who were involved in major medical decision-making for the patient, to either a DA intervention or usual care. The primary outcome was informed decision-making, assessed through measures of knowledge, decisional conflict, decision self-efficacy, and preparation for decision-making. Secondary outcomes included patients' uptake of genetic counseling and testing, participants' CRC screening behavior, healthy lifestyle scores, anxiety and depression levels, quality of life, and satisfaction with the intervention. Data were collected at baseline (T0), after the intervention (T1), and 3 months after the baseline survey (T2). The DA intervention and outcome assessments at T1 and T2 were delivered via WeChat. The effects of the intervention were analyzed using generalized estimating equation models. RESULTS Statistically significant improvements were observed in knowledge (T1: β=2.049, P<.001; T2: β=3.317, P<.001), decisional conflict (T1: β=-11.660, P<.001; T2: β=-17.587, P<.001), and decision self-efficacy (T1: β=15.353, P<.001; T2: β=22.337, P<.001) in the DA group compared with the usual care group at both T1 and T2. Additionally, the DA group showed significantly greater improvement in processed and red meat intake (β=-1.494, P<.001) at T1 and in healthy lifestyle scores (β=1.073, P=.03) at T2. No differences were found between the groups for other outcomes. CONCLUSIONS A DA tool may be a safe, effective, and resource-efficient approach to facilitate informed decision-making about genetic testing. However, the current DA tool requires optimization and further evaluation-for example, by leveraging more advanced technology than WeChat to develop a simpler and more intelligent DA system. TRIAL REGISTRATION Chinese Clinical Trial Registry ChiCTR2100048051; https://www.chictr.org.cn/showproj.html?proj=129054.
Collapse
Affiliation(s)
- Huanhuan Li
- School of Nursing, Jilin University, Changchun, Jilin, China
- School of Nursing, Anhui Medical University, Hefei, Anhui, China
| | - Yanjie Zhao
- School of Nursing, Jilin University, Changchun, Jilin, China
- School of Nursing, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Wei Li
- Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Wenxia Wang
- School of Nursing, Jilin University, Changchun, Jilin, China
- School of Nursing, Peking University, Beijing, China
| | - Shengze Zhi
- School of Nursing, Jilin University, Changchun, Jilin, China
| | - Yifan Wu
- School of Nursing, Jilin University, Changchun, Jilin, China
| | - Qiqing Zhong
- School of Nursing, Jilin University, Changchun, Jilin, China
| | - Rui Wang
- School of Nursing, Jilin University, Changchun, Jilin, China
| | - Jiao Sun
- School of Nursing, Jilin University, Changchun, Jilin, China
| |
Collapse
|
2
|
Cole JJ, Sellitto AD, Baratta LR, Huecker JB, Balls-Berry JJE, Gurnett CA. Social Determinants of Genetics Referral and Completion Rates Among Pediatric Neurology Patients. Pediatr Neurol 2025; 165:78-86. [PMID: 39970807 PMCID: PMC11911075 DOI: 10.1016/j.pediatrneurol.2025.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 05/18/2024] [Accepted: 01/21/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND To investigate clinical, social, and systems-level determinants predictive of genetics clinic referral and completion of genetics clinic visits among pediatric neurology patients. METHODS Electronic health record (EHR) data were extracted from pediatric patients (0-18 years) evaluated in pediatric neurology clinics at a single tertiary care institution between July 2018 and January 2020. Referral and referral completion rates to genetics clinics were compared among non-Hispanic single- or multiracial Black (Black) versus non-Hispanic White (White) patients using bivariablee analysis. Other ethnoracial identities were excluded due to small numbers. Variables associated with genetics clinic referral and visit completion were identified using logistic regressions. RESULTS In a cohort of 11,371 pediatric neurology patients, 304 were referred to genetics clinic and 229 (75.3%) completed genetics clinic visits. In multivariable analyses of Black and White patients (n = 10,601), genetics clinic referral rates did not differ by ethnoracial identity but were associated with younger age, rurality, neurodevelopmental disorder diagnosis, number of neurology clinic visits, and provider type. Genetics clinic visit completion rates were associated with number of neurology clinic visits and ethnoracial identity, with White patients twice as likely as Black patients to complete the visit (adjusted odds ratio=2.18; 95% confidence interval 1.06-4.48). CONCLUSIONS Although no disparity in genetics clinic referral rates was identified, White patients were twice as likely as Black patients to complete a genetics clinic visit after referral. Further work is needed to determine whether this is due to systemic/structural racism, differences in attitudes toward genetics, or other factors.
Collapse
Affiliation(s)
- Jordan J Cole
- Washington University School of Medicine in St. Louis, Department of Neurology, St. Louis, Missouri; University of Colorado Anschutz Medical Campus, Department of Pediatrics, Aurora, Colorado; Children's Hospital Colorado, Pediatric Neuroscience Institute, Aurora, Colorado.
| | - Angela D Sellitto
- Washington University School of Medicine in St. Louis, Department of Neurology, St. Louis, Missouri
| | - Laura Rosa Baratta
- Washington University School of Medicine in St. Louis, Institute for Informatics, Data Science, Biostatistics, St. Louis, Missouri
| | - Julia B Huecker
- Washington University School of Medicine in St. Louis, Institute for Informatics, Data Science, Biostatistics, St. Louis, Missouri
| | - Joyce Joy E Balls-Berry
- Washington University School of Medicine in St. Louis, Department of Neurology, St. Louis, Missouri
| | - Christina A Gurnett
- Washington University School of Medicine in St. Louis, Department of Neurology, St. Louis, Missouri
| |
Collapse
|
3
|
Rustgi SD, Soddano J, Ingram M, Hampel H, Hur C, Kastrinos F. Cost-Effectiveness of Lynch Syndrome Identification Strategies in Individuals with Colorectal Cancer and the Impact on At-Risk Relatives. Clin Gastroenterol Hepatol 2025:S1542-3565(25)00144-2. [PMID: 40010417 DOI: 10.1016/j.cgh.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/23/2024] [Accepted: 01/07/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND & AIMS Universal screening for Lynch syndrome (LS) is recommended for all patients diagnosed with colorectal cancer (CRC). A benefit of LS screening in CRC is cascade testing (CT), whereby at-risk relatives are tested for the familial pathogenic LS variant and undergo intensive surveillance for CRC prevention/early detection if identified with LS. There is not yet universal uptake of CT; we quantify the impact on CRC-related outcomes in first-degree relatives (FDRs). METHODS We developed a microsimulation model to quantify the impact of CT on CRC incidence and mortality in FDRs (parents, siblings, children) of individuals with CRC screened for LS. For FDRs, the primary outcome was the number of CRC cases and CRC-related deaths, by age of relative; secondary outcomes included life-years gained, quality-adjusted life-years, number of colonoscopies, and costs associated with CT, surveillance, and cancer care. RESULTS With CT for all eligible FDRs, we estimate 61.0% decrease in CRC cases and 78.5% decrease in CRC mortality. Although CT led to an average 11 more lifetime colonoscopies, there was modest increase in life-years gained and quality-adjusted life-years and decreased costs because of savings from cancer treatment. CONCLUSIONS This model quantifies the benefits of CT for at-risk FDRs of newly identified individuals with CRC and LS. The decrease in CRC incidence across generations can be used to facilitate discussions with relatives to improve uptake of CT. Further studies to optimize the uptake of CT are paramount to decrease risk of CRC in LS.
Collapse
Affiliation(s)
- Sheila D Rustgi
- Division of Digestive and Liver Diseases, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Josephine Soddano
- Division of Digestive and Liver Diseases, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Myles Ingram
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Heather Hampel
- Division of Clinical Cancer Genomics, Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California
| | - Chin Hur
- Division of Digestive and Liver Diseases, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Fay Kastrinos
- Division of Digestive and Liver Diseases, Columbia University Irving Medical Center, New York, New York; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York.
| |
Collapse
|
4
|
Sears E, Dahlquist J, Stayman S, Ko C, Konnick EQ, Cole A, Zhang Y, Kohn M, Henderson V, Knerr S. Feasibility of using patient navigation to improve identification of hereditary cancer syndromes in newly diagnosed patients with colorectal cancer. Genet Med 2025; 27:101372. [PMID: 39927451 DOI: 10.1016/j.gim.2025.101372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/11/2025] Open
Abstract
PURPOSE Germline genetic testing to identify hereditary cancer syndromes in patients newly diagnosed with colorectal cancer (CRC) carries substantial benefits. We examined the feasibility of using patient navigation, an evidence-based approach to reduce structural barriers to recommended care, to improve test completion by increasing pretest counseling attendance. METHODS We conducted key informant interviews with representatives from organizations providing cancer care to CRC patients. Interviews included questions derived from the Consolidated Framework for Implementation Research, which delineates barriers and facilitators to implementing evidence-based practices. We used an inductive-deductive coding approach to identify themes related to program feasibility. RESULTS We interviewed 19 participants across 13 organizations. Key feasibility barriers included funding to implement and sustain a navigation program, staffing and supervising the navigator role, health information technology needs, gaining administrators' buy-in, and evolving genetic service delivery models. Participants suggested multiple strategies to address implementation barriers, but most would prefer other approaches to improve genetic test completion over implementing a genomics-focused patient navigation program. CONCLUSION Stakeholders across a range of health care organizations saw limited value in improving the identification of hereditary CRC syndromes by implementing a program designed to increase pretest genetic counseling attendance. The need to scale up genetic testing has shifted interest toward delivery models better integrated in established care pathways, requiring fewer resources and providing broader reach.
Collapse
Affiliation(s)
- Emma Sears
- Institute for Public Health Genetics, University of Washington School of Public Health, Seattle, WA
| | - Jacky Dahlquist
- Department of Health Systems and Population Health, University of Washington School of Public Health, Seattle, WA
| | - Sarah Stayman
- Department of Health Systems and Population Health, University of Washington School of Public Health, Seattle, WA
| | - Cynthia Ko
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Eric Q Konnick
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA
| | - Allison Cole
- Department of Family Medicine, University of Washington School of Medicine, Seattle, WA
| | - Ying Zhang
- Department of Family Medicine, University of Washington School of Medicine, Seattle, WA
| | - Marlana Kohn
- Department of Health Systems and Population Health, University of Washington School of Public Health, Seattle, WA
| | - Vida Henderson
- Public Health Sciences Division, Fred Hutch Cancer Center, Seattle, WA
| | - Sarah Knerr
- Department of Health Systems and Population Health, University of Washington School of Public Health, Seattle, WA.
| |
Collapse
|
5
|
Gilbert KM, McLaughlin HM, Farmer JR, Ong MS. Disparities in Genetic Testing for Inborn Errors of Immunity. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2025; 13:388-395.e3. [PMID: 39579980 PMCID: PMC11807750 DOI: 10.1016/j.jaip.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/30/2024] [Accepted: 11/13/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND Inequities in genetic testing have been documented in a range of diseases, and no-charge genetic testing programs have been proposed as a means to enhance access. However, no studies have examined disparities in genetic testing for inborn errors of immunity (IEI) and the impact of no-charge programs on testing equity. OBJECTIVE To examine socioeconomic, geographic, and racial disparities in the uptake of genetic testing for IEI in the United States and the impact of a no-charge sponsored program on testing equity. METHODS This was a retrospective cohort analysis of (1) a national claims database capturing individuals with IEI (n = 18,603), and (2) data from a clinical genetic testing laboratory capturing patients with IEI participating in a no-charge sponsored program (n = 6,681) and a non-sponsored program (n = 29,579) for IEI genetic testing. RESULTS Among patients with IEI captured in the claims database, those residing in areas of greater deprivation (odds ratio [OR] = 0.95; 95% CI, 0.92-0.98), rural areas (OR = 0.82; 95% CI, 0.71-0.96), and non-White neighborhoods (OR = 0.89, 95% CI 0.81-0.98) were less likely to undergo genetic testing. Participants in the sponsored IEI genetic testing program lived in areas of greater deprivation compared with the non-sponsored program (median, 46 vs 42; P < .001). However, historically excluded racial groups were underrepresented in both the sponsored and non-sponsored programs relative to disease burden. CONCLUSIONS We found significant disparities in genetic testing for IEI. Although eliminating the financial barriers to testing reduced socioeconomic disparities in genetic testing for IEI, racial disparities persisted. Further research is needed to address barriers to testing among underserved populations.
Collapse
Affiliation(s)
- Karen M Gilbert
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Boston, Mass.
| | | | - Jocelyn R Farmer
- Clinical Immunodeficiency Program of Beth Israel Lahey Health, Division of Allergy and Immunology, Lahey Hospital and Medical Center, Burlington, Mass
| | - Mei-Sing Ong
- Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Mass
| |
Collapse
|
6
|
Rivera Rivera JN, Snir M, Simmons E, Schmidlen T, Sholeh M, Maconi ML, Geiss C, Fulton H, Barton L, Gonzalez BD, Permuth J, Vadaparampil S. Developing and Assessing a Scalable Digital Health Tool for Pretest Genetic Education in Patients With Early-Onset Colorectal Cancer: Mixed Methods Design. JMIR Cancer 2025; 11:e59464. [PMID: 39819811 PMCID: PMC11786131 DOI: 10.2196/59464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 08/29/2024] [Accepted: 12/16/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND National guidelines recommend germline genetic testing (GT) for all patients with early-onset colorectal cancer. With recent advances in targeted therapies and GT, these guidelines are expected to expand to include broader groups of patients with colorectal cancer. However, there is a shortage of genetic professionals to provide the necessary education and support for informed consent. As such, there is a pressing need to identify alternative approaches to facilitate and expedite access to GT. OBJECTIVE This study describes the development of a pretest education intervention, Nest-CRC, to facilitate the uptake of germline GT among patients with early-onset colorectal cancer. Patients with early-onset colorectal cancer and health care providers reviewed Nest-CRC, and their reactions and recommendations were captured using a nested mixed methods approach. METHODS Using the learner verification approach, we conducted 2 sequential phases of surveys and interviews with English- and Spanish-speaking patients with early-onset colorectal cancer and health care providers. The surveys assessed participants' experiences with genetic services and provided immediate feedback on the Nest-CRC genetic education modules. Semistructured interviews evaluated participants' perceptions of self-efficacy, attraction, comprehension, cultural acceptability, and usability of Nest-CRC. Survey data were analyzed using descriptive statistics (mean, median, and proportions), while interview data were analyzed through line-by-line coding of the transcribed interviews. After each phase, Nest-CRC was refined based on participants' recommendations. RESULTS A total of 52 participants, including 39 patients with early-onset colorectal cancer and 13 providers, participated in the study. Of these, 19 patients and 6 providers participated in phase 1 (N=25), and 20 patients and 7 providers participated in phase 2 (N=27). Most participants (phase 1: 23/25, 92%, to 25/25, 100%; phase 2: 24/27, 89%, to 27/27, 100%) agreed that each of the 5 education modules was easy to understand and helpful; 13 patients reported no history of GT, with 11 (85%) expressing interest in GT and 2 (15%) remaining unsure after completing Nest-CRC. Participants reported that Nest-CRC provided sufficient information to help them decide about GT. The tool was deemed acceptable by individuals from diverse backgrounds, and participants found it visually attractive, easy to comprehend, and user-friendly. CONCLUSIONS The findings revealed that Nest-CRC is a promising strategy for facilitating pretest education and promoting GT. Nest-CRC has been refined based on participant recommendations and will be re-evaluated.
Collapse
Affiliation(s)
- Jessica N Rivera Rivera
- Healthcare Delivery Research Network, MedStar Health Research Institute, Washington, DC, United States
| | - Moran Snir
- Nest Genomics, New York, NY, United States
| | | | | | - Misha Sholeh
- Non-Therapeutic Research Office, Moffitt Cancer Center, Tampa, FL, United States
| | - Melinda Leigh Maconi
- Participant Research, Interventions, and Measurement Core, Moffitt Cancer Center, Tampa, FL, United States
| | - Carley Geiss
- Participant Research, Interventions, and Measurement Core, Moffitt Cancer Center, Tampa, FL, United States
| | - Hayden Fulton
- Participant Research, Interventions, and Measurement Core, Moffitt Cancer Center, Tampa, FL, United States
| | - Laura Barton
- Department of Pathology, Moffitt Cancer Center, Tampa, FL, United States
| | - Brian D Gonzalez
- Research Diversity and Workforce Development, Moffitt Cancer Center, Tampa, FL, United States
| | - Jennifer Permuth
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Susan Vadaparampil
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
7
|
Li C, Chen T, Chen H, Zhang B, Sun B, Zhou P, Li Q, Chen W. Temporal Trends in Colorectal Cancer Incidence and Case Numbers among Individuals Aged 45-49 in the US During 2001-2019. Cancer Control 2025; 32:10732748251327715. [PMID: 40183344 PMCID: PMC11970092 DOI: 10.1177/10732748251327715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/19/2025] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
Objective: This study aimed to update the temporal trends for the incidences and case numbers of colorectal cancer (CRC) among individuals aged 45-49 in the US from 2001 to 2019.Methods: Patients were obtained from the National Program of Cancer Registries and Surveillance, Epidemiology and End Results Program (NPCR-SEER) database. Their age-adjusted incidence rates (AAIR) were calculated using the SEER*Stat software.Results: As high as 48.4% (125 604 cases) of the 259 700 early-onset CRC were diagnosed in individuals aged 45-49. Of these, 54.2% were males, and 40.7% were located in the rectum. Adenocarcinoma accounted for 93.9%, 96.5%, and 84.6% of proximal, distal colon, and rectal cancers, respectively. The incidences of proximal colon adenocarcinoma showed a significant increase, with an average annual percentage change (APC) of 0.7 from 2010 to 2019, while the case numbers remained stable from 2001 to 2019. In contrast, distal colon adenocarcinoma displayed increased incidences at an APC of 1.3 and an average increase of 17 cases annually over the study period. Rectal adenocarcinoma showed more rapid increases in incidence, with an average APC of 1.6 and an average increase of 27 cases per year. These rising incidences were predominately observed in non-Hispanic whites (NHWs). Conversely, non-Hispanic black (NHB) females showed decreased incidences of proximal and distal colon adenocarcinoma. Additionally, the incidences and case numbers for carcinoids significantly increased in the rectum but not in the colon.Conclusions: This study reveals distinct patterns of temporal trends in CRC incidences and case numbers among individuals aged 45-49. Further research is necessary to understand the underlying causes of the differences and to develop more effective preventive strategies to reduce the incidence of early-onset CRC.
Collapse
Affiliation(s)
- Chunmei Li
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Science, Hangzhou, China
- Post-Graduate Training, Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Tianle Chen
- Department of Mathematics, University of Wisconsin-Madison, Madison, WI, USA
| | - Huimin Chen
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Science, Hangzhou, China
- Post-Graduate Training, Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Bo Zhang
- Department of Colorectal Surgery, Jinhua People’s Hospital, Jinhua, China
| | - Bing Sun
- Department of Gastrointestinal Surgery, Haining People’s Hospital, Jiaxing, China
| | - Pengyang Zhou
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Science, Hangzhou, China
| | - Qiken Li
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Science, Hangzhou, China
| | - Weiping Chen
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Science, Hangzhou, China
| |
Collapse
|
8
|
Hill RC, Wang Y, Shaikh B, Lipner SR. Future research directions regarding safety of spironolactone for dermatologic conditions. J Am Acad Dermatol 2024; 91:e173-e174. [PMID: 39154749 DOI: 10.1016/j.jaad.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/29/2024] [Accepted: 08/01/2024] [Indexed: 08/20/2024]
Affiliation(s)
| | - Yu Wang
- Department of Dermatology, Wake Forest School of Medicine, Wake Forest, North Carolina
| | - Bilal Shaikh
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York, New York
| | - Shari R Lipner
- Department of Dermatology, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
9
|
Lavingia V, Sardana S, Khanderia M, Bisht N, Patel A, Koyyala VPB, Sheth H, Ramaswamy A, Singh A, deSouza A, Jain SB, Mahajan M, Gohel S, Parikh A, Brown G, Sirohi B. Localized Rectal Cancer: Indian Consensus and Guidelines. Indian J Med Paediatr Oncol 2024; 45:461-480. [DOI: 10.1055/s-0043-1777865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
AbstractThe rising incidence of colorectal cancer (CRC) in India, particularly the prevalence of rectal cancer over colon cancer (0.7:1), has been a growing concern in recent decades; especially notable is the trend of increasing cases among young CRC patients. Given the diverse treatment approaches for rectal cancer globally and the varying economic capacities of patients in low to middle-income countries (LMICs) like India, it is essential to establish consensus guidelines that are specifically tailored to meet the needs of these patients. To achieve this, a panel comprising 30 eminent rectal cancer experts convened to conduct a comprehensive and impartial evaluation of existing practices and recent advancements in the field. Through meticulous scrutiny of published literature and a consensus-building process that involved voting on pertinent questions, the panel formulated management strategies. These recommendations are the result of a rigorous, evidence-based process and encapsulate the collective wisdom and judgment of leading authorities in the field.
Collapse
Affiliation(s)
- Viraj Lavingia
- Department of Medical Oncology, HCG Cancer Center, Ahmedabad, Gujarat, India
| | - Shefali Sardana
- Department of Medical Oncology, Max Institute of Cancer Care, Max Superspeciality Hospital, New Delhi, India
| | - Mansi Khanderia
- Department of Medical Oncology, SPARSH Hospitals, Bangalore, Karnataka, India
| | - Niharika Bisht
- Department of Radiation Oncology, Army Hospital Research and Referral, New Delhi, India
| | - Amol Patel
- Department of Medical Oncology, Indian Naval Hospital Ship Asvini, Mumbai, Maharashtra, India
| | | | - Harsh Sheth
- Department of Advanced Genomic Technologies Division, FRIGE Institute of Human Genetics, Ahmedabad, Gujarat, India
| | - Anant Ramaswamy
- Department of Medical Oncology, Tata Memorial Centre (HBNI), Mumbai, Maharashtra, India
| | - Ashish Singh
- Department of Medical Oncology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Ashwin deSouza
- Department of Surgical Oncology, Tata Memorial Centre and Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sneha Bothra Jain
- Department of Medical Oncology, Mittal Institute of Medical Sciences, Bhilai, Chhattisgarh, India
| | - Mukta Mahajan
- Department of Radiodiagnosis, Apollo Proton Cancer Centre, Chennai, Tamil Nadu, India
| | - Shruti Gohel
- Department of Medical Oncology, HCG Cancer Centre, Ahmedabad, Gujarat, India
| | - Aparna Parikh
- Department of Medical Oncology, Mass General Cancer Centre, Boston, United States
| | - Gina Brown
- Department of Gastrointestinal Cancer Imaging, Imperial College, London, United Kingdom
| | | |
Collapse
|
10
|
Lau-Min KS, Rochester S, Grabill M, Long JM, McKenna DB, Powers J, Bracy D, Boisseau L, Gabriel P, Oyer R, Domchek SM, Rendle KA, Nathanson KL, Katona BW. Pilot implementation study of a default genetic referral process for patients with early-onset colorectal cancer. GENETICS IN MEDICINE OPEN 2024; 3:101902. [PMID: 39896148 PMCID: PMC11780105 DOI: 10.1016/j.gimo.2024.101902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 02/04/2025]
Abstract
Purpose Early-onset colorectal cancer (CRC) diagnosed under age 50 is increasing at alarming rates, with >75% of early-onset cases occurring in patients between 40 and 49 years old. Germline genetic risk evaluations are key to delivering high-quality care to these patients. Methods We conducted a single-arm pilot implementation study of a default genetic referral process for patients diagnosed with CRC between ages 40 and 49 at 5 hospitals in an academic health system. A research coordinator notified patients and their oncologists of their eligibility for a default genetic referral, after which all patients who did not opt out were referred for genetic counseling, testing, and result disclosure as per usual care. The primary outcome was the genetic referral rate; secondary outcomes included the percentage of eligible patients who were scheduled for a genetic evaluation, completed genetic counseling, and underwent testing within 3 months of the initial referral. We conducted semistructured exit interviews with a subset of patients and oncologists to elicit feedback on the intervention. Results We included 53 patients, of whom 49 (92%) were referred to genetics, 38 (72%) were scheduled, 22 (42%) completed genetic counseling, and 13 (25%) underwent testing within 3 months of the initial referral. In exit interviews (n = 10 patients and 10 oncologists), participants reported finding the default genetic referral process acceptable and feasible to implement. Conclusion A default genetic referral process is acceptable, feasible, and associated with a high referral rate for patients with early-onset CRC; however, subsequent scheduling, evaluation, and testing rates remain suboptimal.
Collapse
Affiliation(s)
- Kelsey S. Lau-Min
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Shavon Rochester
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Megan Grabill
- Department of Family Medicine and Community Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jessica M. Long
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Danielle B. McKenna
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Jacqueline Powers
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Danny Bracy
- Department of Family Medicine and Community Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Leland Boisseau
- Information Services Applications, Penn Medicine, University of Pennsylvania, Philadelphia, PA
| | - Peter Gabriel
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Randall Oyer
- Ann B. Barshinger Cancer Institute, Penn Medicine at Lancaster General Health, Lancaster, PA
| | - Susan M. Domchek
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Katharine A. Rendle
- Department of Family Medicine and Community Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Katherine L. Nathanson
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Bryson W. Katona
- Division of Gastroenterology and Hepatology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
11
|
Chai TS, Rodgers-Fouche LH, Walls JO, Mattia AR, Chung DC. Real-world genetic testing outcomes of pan-cancer testing for mismatch repair deficiency. Cancer 2024; 130:3888-3893. [PMID: 39024159 DOI: 10.1002/cncr.35473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/17/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND In 2017, the Food and Drug Administration approved pembrolizumab for treatment of any mismatch repair-deficient (dMMR) tumor making MMR immunohistochemistry (IHC) testing beneficial for all tumor types. For the first time, MMR IHC was not performed exclusively to screen for Lynch syndrome (LS). METHODS In this study, all MMR IHC reports issued between 2017 and 2021 at an academic hospital were reviewed and completion of genetic testing was determined through chart review. Colorectal cancers (CRCs), endometrial cancers (ECs), and noncancerous lesions were excluded. RESULTS Between 2017 and 2021, MMR IHC was completed in 1939 patients with a malignancy other than CRC or EC. Absent or weak staining for at least one MMR protein was detected in 115 (5.9%) patients and 59 (51%) of those completed germline genetic testing. Overall, the identification rate of LS in this cohort was 0.72%, which is similar to the rate in our previously reported CRC and EC universal screening cohort. A diagnosis of LS was most commonly made in patients with dMMR brain (18.75%) and small intestinal cancers (10.20%). Five additional patients were found to carry a pathogenic variant in a non-LS gene. CONCLUSIONS Pan-cancer MMR testing for pembrolizumab consideration can identify LS cases at a rate similar to universal CRC and EC screening programs. A persistent challenge is subsequent uptake of genetic testing. MMR testing should be prioritized in brain and small intestinal tumors, and multigene panel testing is recommended in patients with dMMR, as unexpected pathogenic variants in non-LS genes were found as frequently as LS gene variants.
Collapse
Affiliation(s)
- Teresa S Chai
- Center for Cancer Risk Assessment, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Linda H Rodgers-Fouche
- Center for Cancer Risk Assessment, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jenna O Walls
- Center for Cancer Risk Assessment, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Anthony R Mattia
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Daniel C Chung
- Center for Cancer Risk Assessment, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Bonini KE, Smith HS, Bonkowski ES, Berkman BE, Jamal L. Modern Family: An Ethical Justification for System-Led Contact of Relatives Eligible for Cascade Screening in the United States. Public Health Genomics 2024; 28:19-33. [PMID: 39522512 DOI: 10.1159/000541301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 09/03/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Though genomic science has rapidly advanced, efforts to demonstrate the population-level utility of genomics have been slow to follow. It has long been argued that the family is an important unit of significance in genomics, yet it has been challenging to address this in clinical care. This is apparent in how hospital administrators and clinicians in the United States typically approach cascade screening, the process of notifying and offering genetic testing to at-risk relatives of a patient with a hereditary condition. The most common notification approach is proband-led contact, in which the index patient is responsible for communicating a health risk to their relatives. This model has been associated with suboptimal outcomes. In contrast, recent research has shown that system-led contact, in which healthcare or public health institutions initiate communication to relatives with the proband's consent, has been associated with increased clinical utility and acceptability. SUMMARY With the needs of hospital administrators and clinicians in mind, we revisit normative questions about the appropriate way to notify relatives about their potentially elevated risk of developing an actionable disease. We review evidence demonstrating that system-led direct contact of relatives is feasible and acceptable. We further argue that system-led contact of relatives eligible for cascade screening is ethically justified if these programs are designed with public input, have an opt-out provision, and are implemented for conditions that meet specific criteria which we propose in this article. KEY MESSAGES In this article, we emphasize the usefulness of public health ethics frameworks to inform the design of system-led contact programs. Beyond this, we make the case that such programs are necessary to realize the population utility of genomic medicine equitably.
Collapse
Affiliation(s)
- Katherine E Bonini
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Hadley Stevens Smith
- Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, Massachusetts, USA
- Center for Bioethics, Harvard Medical School, Boston, Massachusetts, USA
| | - Emily S Bonkowski
- Institute for Public Health Genetics, University of Washington School of Public Health, Seattle, Washington, USA
- Center for Pediatric Neurological Disease Research, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Benjamin E Berkman
- Department of Bioethics, National Institutes of Health, Bethesda, Maryland, USA
- National Human Genome Research Institute, Bethesda, Maryland, USA
| | - Leila Jamal
- Department of Bioethics, National Institutes of Health, Bethesda, Maryland, USA
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
13
|
Meernik C, Wang F, Raveendran Y, Green MF, Check DK, Bosworth HB, Sutton LM, Strickler JH, Akinyemiju TF. Association of Race and Ethnicity with Genomic Testing at a Comprehensive Cancer Center in North Carolina. CANCER RESEARCH COMMUNICATIONS 2024; 4:2968-2975. [PMID: 39440958 PMCID: PMC11570879 DOI: 10.1158/2767-9764.crc-24-0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/28/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
SIGNIFICANCE Non-Hispanic Black patients diagnosed with prostate cancer between 2014 and 2019 and treated at a comprehensive cancer center were less likely to use tumor-specific genomic testing compared with non-Hispanic White patients. Disparities in the use of precision oncology technologies should be monitored and addressed to ensure equitable cancer care.
Collapse
Affiliation(s)
- Clare Meernik
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
| | - Frances Wang
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina
| | | | - Michelle F. Green
- Department of Pathology, Duke University School of Medicine, Durham, North Carolina
| | - Devon K. Check
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
| | - Hayden B. Bosworth
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
- Center of Innovation to Accelerate Discovery and Practice Transformation, Durham Veterans Affairs Medical Center, Durham, North Carolina
- Division of General Internal Medicine, Department of Medicine, Duke University Medical Center, Durham, North Carolina
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina
- Duke University School of Nursing, Duke University School of Medicine, Durham, North Carolina
| | - Linda M. Sutton
- Duke Cancer Network, Duke Cancer Institute, Durham, North Carolina
| | - John H. Strickler
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina
| | - Tomi F. Akinyemiju
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
14
|
Chambers DA, Goddard KAB. Advancing Implementation Science in Cancer Genomics: Progressing from Discovery to Population Health Benefit. Public Health Genomics 2024; 27:161-167. [PMID: 39374589 DOI: 10.1159/000541577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024] Open
Affiliation(s)
- David A Chambers
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| | - Katrina A B Goddard
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
15
|
Kulchak Rahm A, Wolfinger T, Salvati ZM, Schneider JL, Cragun D. Development, Evaluation, and User Testing of a Decision-Making Toolkit to Promote Organizations to Implement Universal Tumor Screening for Lynch Syndrome. Public Health Genomics 2024; 27:136-149. [PMID: 39159623 DOI: 10.1159/000540943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024] Open
Abstract
INTRODUCTION The Implementing Universal Lynch Syndrome Screening (IMPULSS) study explained institutional variation in universal tumor screening (UTS) with the goal of identifying ways to aid organizational decision-makers in implementing and optimizing Lynch syndrome UTS programs. METHODS After applying the Consolidated Framework for Implementation Research (CFIR 1.0) to analyze interviews with 66 stakeholders across 9 healthcare systems to develop a toolkit for implementation, we adapted the International Patient Decision Aid Standards (IPDAS) to assess toolkit potential to aid decision-making consistent with organizational values. We then conducted user testing with two experienced and four non-experienced implementers of UTS to improve the content and functionality of the toolkit and assess its acceptability and appropriateness. RESULTS Toolkit components were organized to address findings related to CFIR 1.0 constructs of evidence strength and quality, relative advantage, cost, engaging, planning, executing, and reflecting and evaluating. A home page was added to direct users to different sections based on whether they are deciding to implement UTS, planning for implementation, improving an existing UTS program, or considering a different approach to identify patients with Lynch syndrome. Upon initial evaluation, 31 of 64 IPDAS criteria were met by the original toolkit. All users rated the toolkit as acceptable and appropriate for assisting organizational decision-making and identified multiple areas for improvement. Numerous iterative changes were made to the toolkit, resulting in meeting 17 of the previously unmet IPDAS criteria. CONCLUSION We demonstrate the rigorous development of a toolkit guided by the CFIR and show how user testing helped improve the toolkit to ensure it is acceptable, appropriate, and meets most IPDAS criteria relevant to organizational values-based decision-making.
Collapse
Affiliation(s)
| | | | - Zachary M Salvati
- Department of Genomic Health, Geisinger, Danville, Pennsylvania, USA
| | | | | |
Collapse
|
16
|
Morgan KM, Campbell-Salome G, Walters NL, Betts MN, Brangan A, Johns A, Kirchner HL, Lindsey-Mills Z, McGowan MP, Tricou EP, Rahm AK, Sturm AC, Jones LK. Innovative Implementation Strategies for Familial Hypercholesterolemia Cascade Testing: The Impact of Genetic Counseling. J Pers Med 2024; 14:841. [PMID: 39202032 PMCID: PMC11355397 DOI: 10.3390/jpm14080841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
The IMPACT-FH study implemented strategies (packet, chatbot, direct contact) to promote family member cascade testing for familial hypercholesterolemia (FH). We evaluated the impact of genetic counseling (GC) on medical outcomes, strategy selection, and cascade testing. Probands (i.e., patients with FH) were recommended to complete GC and select sharing strategies. Comparisons were performed for both medical outcomes and strategy selection between probands with or without GC. GEE models for Poisson regression were used to examine the relationship between proband GC completion and first-degree relative (FDR) cascade testing. Overall, 46.3% (81/175) of probands completed GC. Probands with GC had a median LDL-C reduction of -13.0 mg/dL (-61.0, 4.0) versus -1.0 mg/dL (-16.0, 17.0) in probands without GC (p = 0.0054). Probands with and without GC selected sharing strategies for 65.3% and 40.3% of FDRs, respectively (p < 0.0001). Similarly, 27.1% of FDRs of probands with GC completed cascade testing, while 12.0% of FDRs of probands without GC completed testing (p = 0.0043). Direct contact was selected for 47 relatives in total and completed for 39, leading to the detection of 18 relatives with FH. Proband GC was associated with improved medical outcomes and increased FDR cascade testing. Direct contact effectively identified FH cases for the subset who participated.
Collapse
Affiliation(s)
- Kelly M. Morgan
- Department of Genomic Health, Research Institute, Geisinger, 100 N. Academy Avenue, Danville, PA 17922, USA; (G.C.-S.); (N.L.W.); (A.B.); (Z.L.-M.); (A.C.S.)
| | - Gemme Campbell-Salome
- Department of Genomic Health, Research Institute, Geisinger, 100 N. Academy Avenue, Danville, PA 17922, USA; (G.C.-S.); (N.L.W.); (A.B.); (Z.L.-M.); (A.C.S.)
- Department of Population Health Sciences, Research Institute, Geisinger, 100 N. Academy Avenue, Danville, PA 17922, USA;
| | - Nicole L. Walters
- Department of Genomic Health, Research Institute, Geisinger, 100 N. Academy Avenue, Danville, PA 17922, USA; (G.C.-S.); (N.L.W.); (A.B.); (Z.L.-M.); (A.C.S.)
| | - Megan N. Betts
- WellSpan Health, 605 S. George Street, York, PA 17401, USA;
| | - Andrew Brangan
- Department of Genomic Health, Research Institute, Geisinger, 100 N. Academy Avenue, Danville, PA 17922, USA; (G.C.-S.); (N.L.W.); (A.B.); (Z.L.-M.); (A.C.S.)
| | - Alicia Johns
- Biostatistics Core, Research Institute, Geisinger, 100 N. Academy Avenue, Danville, PA 17922, USA;
| | - H. Lester Kirchner
- Department of Population Health Sciences, Research Institute, Geisinger, 100 N. Academy Avenue, Danville, PA 17922, USA;
| | - Zoe Lindsey-Mills
- Department of Genomic Health, Research Institute, Geisinger, 100 N. Academy Avenue, Danville, PA 17922, USA; (G.C.-S.); (N.L.W.); (A.B.); (Z.L.-M.); (A.C.S.)
| | - Mary P. McGowan
- Family Heart Foundation, 605 E. Colorado Blvd Ste 180, Pasadena, CA 91101, USA (E.P.T.)
| | - Eric P. Tricou
- Family Heart Foundation, 605 E. Colorado Blvd Ste 180, Pasadena, CA 91101, USA (E.P.T.)
| | - Alanna Kulchak Rahm
- Department of Genomic Health, Research Institute, Geisinger, 100 N. Academy Avenue, Danville, PA 17922, USA; (G.C.-S.); (N.L.W.); (A.B.); (Z.L.-M.); (A.C.S.)
| | - Amy C. Sturm
- Department of Genomic Health, Research Institute, Geisinger, 100 N. Academy Avenue, Danville, PA 17922, USA; (G.C.-S.); (N.L.W.); (A.B.); (Z.L.-M.); (A.C.S.)
- Heart and Vascular Institute, Geisinger, 100 N. Academy Avenue, Danville, PA 17922, USA
- 23andMe, 223 N. Mathilda Avenue, Sunnyvale, CA 94086, USA
| | - Laney K. Jones
- Department of Genomic Health, Research Institute, Geisinger, 100 N. Academy Avenue, Danville, PA 17922, USA; (G.C.-S.); (N.L.W.); (A.B.); (Z.L.-M.); (A.C.S.)
- Heart and Vascular Institute, Geisinger, 100 N. Academy Avenue, Danville, PA 17922, USA
| |
Collapse
|
17
|
Allen CG, Hunt K, Jackson A, Baierl J, McMahon L, Judge D. Applying the R = MC 2 implementation science heuristic to assess the impact of readiness on reach and implementation of a population-wide genomic screening program. J Genet Couns 2024; 33:815-821. [PMID: 37732417 PMCID: PMC10954584 DOI: 10.1002/jgc4.1775] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/30/2023] [Accepted: 08/14/2023] [Indexed: 09/22/2023]
Abstract
Population-wide genomic screening for genes that have high penetrance and clinical actionability enhances the opportunity to identify individuals at risk for developing hereditary conditions. Organizational readiness has been shown to influence the likelihood of successful implementation of complex initiatives such as the integration of population-wide genomic screening in clinical settings. We use the organizational readiness heuristic R = MC2 to better understand three factors that influence readiness for implementation of In Our DNA SC, a population-wide genomic screening program: motivation to implement, general capacity of an organization, and innovation-specific capacities. We then assessed the influence of these readiness factors on implementation outcomes of reach (measured through enrollment rate) and implementation (measured through the number of DNA samples collected). Data were collected pre-implementation and captured during the three-month pilot phase of the In Our DNA SC program. We collected administrative data from the electronic health record and quantitatively captured elements of readiness through surveys distributed to provider champions and clinical administrative champions at the 10 sites implementing the population-wide genomic screening program. We facilitated innovation-specific capacity through training offered at each site, as well as technical assistance through weekly meetings with other implementing sites, and resources available to all staff. Forty percent of provider champions attended training and 80% of administrative champions attended training. An average of 3.7 additional staff were trained at each implementing site. Satisfaction with training positively influenced reach (β = 0.0121, p = 0.0271) but did not impact implementation. Provider engagement (innovation capabilities) was associated with reach (β = 0.0020, p = 0.0251) and clinical administrator engagement was associated with sample collection rate (β = 0.2599, β = 0.038). Readiness to change is considered one of the most important factors in understanding the potential opportunity for implementation. We found that motivation to adopt a population-wide genomic screening program positively impacted the program's reach. The type of champion influenced discrete outcomes, with provider champions positively impacting reach and administrative champions influencing implementation (assessed through sample collection rate). As genomics continues to be integrated into clinical practice, it will be important to understand the contextual factors that influence readiness for implementation and design support throughout the life-course of implementation to ensure the success of large-scale, complex initiatives.
Collapse
Affiliation(s)
- Caitlin G. Allen
- Medical University of South Carolina, Charleston, South Carolina, USA
| | - Kelly Hunt
- Medical University of South Carolina, Charleston, South Carolina, USA
| | - Amy Jackson
- Medical University of South Carolina, Charleston, South Carolina, USA
| | - Joseph Baierl
- Medical University of South Carolina, Charleston, South Carolina, USA
| | - Lori McMahon
- Medical University of South Carolina, Charleston, South Carolina, USA
| | - Daniel Judge
- Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
18
|
Samadder NJ, Gay E, Lindpere V, Bublitz ML, Bandel LA, Armasu SM, Vierkant RA, Ferber MJ, Klee EW, Larson NB, Kruisselbrink TM, Egan JB, Kemppainen JL, Bidwell JS, Anderson JL, McAllister TM, Walker TS, Kunze KL, Golafshar MA, Klint MA, Presutti RJ, Bobo WV, Sekulic A, Summer-Bolster JM, Willman CL, Lazaridis KN. Exome Sequencing Identifies Carriers of the Autosomal Dominant Cancer Predisposition Disorders Beyond Current Practice Guideline Recommendations. JCO Precis Oncol 2024; 8:e2400106. [PMID: 39013133 DOI: 10.1200/po.24.00106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/28/2024] [Accepted: 05/17/2024] [Indexed: 07/18/2024] Open
Abstract
PURPOSE The autosomal dominant cancer predisposition disorders hereditary breast and ovarian cancer (HBOC) and Lynch syndrome (LS) are genetic conditions for which early identification and intervention have a positive effect on the individual and public health. The goals of this study were to determine whether germline genetic screening using exome sequencing could be used to efficiently identify carriers of HBOC and LS. METHODS Participants were recruited from three geographically and racially diverse sites in the United States (Rochester, MN; Phoenix, AZ; Jacksonville, FL). Participants underwent Exome+ sequencing (Helix Inc, San Mateo, CA) and return of results for specific genetic findings: HBOC (BRCA1 and BRCA1) and LS (MLH1, MSH2, MSH6, PMS2, and EPCAM). Chart review was performed to collect demographics and personal and family cancer history. RESULTS To date, 44,306 participants have enrolled in Tapestry. Annotation and interpretation of all variants in genes for HBOC and LS resulted in the identification of 550 carriers (prevalence, 1.24%), which included 387 with HBOC (27.2% BRCA1, 42.8% BRCA2) and 163 with LS (12.3% MSH6, 8.8% PMS2, 4.5% MLH1, 3.8% MSH2, and 0.2% EPCAM). More than half of these participants (52.1%) were newly diagnosed carriers with HBOC and LS. In all, 39.2% of HBOC/LS carriers did not satisfy National Comprehensive Cancer Network (NCCN) criteria for genetic evaluation. NCCN criteria were less commonly met in underrepresented minority populations versus self-reported White race (51.5% v 37.5%, P = .028). CONCLUSION Our results emphasize the need for wider utilization of germline genetic sequencing for enhanced screening and detection of individuals who have LS and HBOC cancer predisposition syndromes.
Collapse
Affiliation(s)
- N Jewel Samadder
- Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Phoenix, AZ
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Comprehensive Cancer Center, Mayo Clinic, Rochester, MN
| | - Emily Gay
- Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
- Genetic Counseling Program, Arizona State University, Tucson, AZ
| | - Vanda Lindpere
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ
| | | | | | | | - Robert A Vierkant
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Matthew J Ferber
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Eric W Klee
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | | | - Jan B Egan
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | - Jessa S Bidwell
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | | | | | - T'Nita S Walker
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Katie L Kunze
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ
| | | | | | - Richard J Presutti
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Family Medicine, Mayo Clinic, Jacksonville, FL
| | - William V Bobo
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Psychiatry & Psychology, Mayo Clinic, Jacksonville, FL
| | - Aleksander Sekulic
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Dermatology, Mayo Clinic, Scottsdale, AZ
| | | | - Cheryl L Willman
- Comprehensive Cancer Center, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Konstantinos N Lazaridis
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Comprehensive Cancer Center, Mayo Clinic, Rochester, MN
- Division of Gastroenterology & Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
19
|
Maoz A, Yurgelun MB. Leveraging Electronic Health Record Data to Understand Gaps Underlying the Underdiagnosis of Lynch Syndrome. JCO Clin Cancer Inform 2024; 8:e2400032. [PMID: 38838279 DOI: 10.1200/cci.24.00032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/09/2024] [Indexed: 06/07/2024] Open
Abstract
Using the electronic health record to address the underdiagnosis of Lynch syndrome.
Collapse
Affiliation(s)
- Asaf Maoz
- Dana-Farber Cancer Institute, Boston, MA
- Brigham & Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Matthew B Yurgelun
- Dana-Farber Cancer Institute, Boston, MA
- Brigham & Women's Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
20
|
Shevach JW, Candelieri-Surette D, Lynch JA, Hubbard RA, Alba PR, Glanz K, Parikh RB, Maxwell KN. Racial Differences in Germline Genetic Testing Completion Among Males With Pancreatic, Breast, or Metastatic Prostate Cancers. J Natl Compr Canc Netw 2024; 22:237-243. [PMID: 38631387 PMCID: PMC11361447 DOI: 10.6004/jnccn.2023.7105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/27/2023] [Indexed: 04/19/2024]
Abstract
BACKGROUND Germline genetic testing is a vital component of guideline-recommended cancer care for males with pancreatic, breast, or metastatic prostate cancers. We sought to determine whether there were racial disparities in germline genetic testing completion in this population. PATIENTS AND METHODS This retrospective cohort study included non-Hispanic White and Black males with incident pancreatic, breast, or metastatic prostate cancers between January 1, 2019, and September 30, 2021. Two nationwide cohorts were examined: (1) commercially insured individuals in an administrative claims database, and (2) Veterans receiving care in the Veterans Health Administration. One-year germline genetic testing rates were estimated by using Kaplan-Meier methods. Cox proportional hazards regression was used to test the association between race and genetic testing completion. Causal mediation analyses were performed to investigate whether socioeconomic variables contributed to associations between race and germline testing. RESULTS Our cohort consisted of 7,894 males (5,142 commercially insured; 2,752 Veterans). One-year testing rates were 18.0% (95% CI, 16.8%-19.2%) in commercially insured individuals and 14.2% (95% CI, 11.5%-15.0%) in Veterans. Black race was associated with a lower hazard of testing among commercially insured individuals (adjusted hazard ratio [aHR], 0.73; 95% CI, 0.58-0.91; P=.005) but not among Veterans (aHR, 0.99; 95% CI, 0.75-1.32; P=.960). In commercially insured individuals, income (aHR, 0.90; 95% CI, 0.86-0.96) and net worth (aHR, 0.92; 95% CI, 0.86-0.98) mediated racial disparities, whereas education (aHR, 0.98; 95% CI, 0.94-1.01) did not. CONCLUSIONS Overall rates of guideline-recommended genetic testing are low in males with pancreatic, breast, or metastatic prostate cancers. Racial disparities in genetic testing among males exist in a commercially insured population, mediated by net worth and household income; these disparities are not seen in the equal-access Veterans Health Administration. Alleviating financial and access barriers may mitigate racial disparities in genetic testing.
Collapse
Affiliation(s)
- Jeffrey W. Shevach
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC
| | | | - Julie A. Lynch
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT
| | - Rebecca A. Hubbard
- Department of Biostatistics, Epidemiology & Informatics, University of Pennsylvania, Philadelphia, PA
| | - Patrick R. Alba
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT
| | - Karen Glanz
- Perelman School of Medicine and School of Nursing, University of Pennsylvania, Philadelphia, PA
| | - Ravi B. Parikh
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kara N. Maxwell
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
21
|
Baldwin A, Copeland J, Azage M, Dratch L, Johnson K, Paul RA, Amado DA, Baer M, Deik A, Elman LB, Guo M, Hamedani AG, Irwin DJ, Lasker A, Orthmann-Murphy J, Quinn CC, Tropea TF, Scherer SS, Shinohara RT, Hamilton RH, Ellis CA. Disparities in Genetic Testing for Neurologic Disorders. Neurology 2024; 102:e209161. [PMID: 38447117 PMCID: PMC11383874 DOI: 10.1212/wnl.0000000000209161] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/01/2023] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Genetic testing is now the standard of care for many neurologic conditions. Health care disparities are unfortunately widespread in the US health care system, but disparities in the utilization of genetic testing for neurologic conditions have not been studied. We tested the hypothesis that access to and results of genetic testing vary according to race, ethnicity, sex, socioeconomic status, and insurance status for adults with neurologic conditions. METHODS We analyzed retrospective data from patients who underwent genetic evaluation and testing through our institution's neurogenetics program. We tested for differences between demographic groups in 3 steps of a genetic evaluation pathway: (1) attending a neurogenetic evaluation, (2) completing genetic testing, and (3) receiving a diagnostic result. We compared patients on this genetic evaluation pathway with the population of all neurology outpatients at our institution, using univariate and multivariable logistic regression analyses. RESULTS Between 2015 and 2022, a total of 128,440 patients were seen in our outpatient neurology clinics and 2,540 patients underwent genetic evaluation. Black patients were less than half as likely as White patients to be evaluated (odds ratio [OR] 0.49, p < 0.001), and this disparity was similar after controlling for other demographic factors in multivariable analysis. Patients from the least wealthy quartile of zip codes were also less likely to be evaluated (OR 0.67, p < 0.001). Among patients who underwent evaluation, there were no disparities in the likelihood of completing genetic testing, nor in the likelihood of a diagnostic result after adjusting for age. Analyses restricted to specific indications for genetic testing supported these findings. DISCUSSION We observed unequal utilization of our clinical neurogenetics program for patients from marginalized and minoritized demographic groups, especially Black patients. Among patients who do undergo evaluation, all groups benefit similarly from genetic testing when it is indicated. Understanding and removing barriers to accessing genetic testing will be essential to health care equity and optimal care for all patients with neurologic disorders.
Collapse
Affiliation(s)
- Aaron Baldwin
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Juliette Copeland
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Meron Azage
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Laynie Dratch
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Kelsey Johnson
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Rachel A Paul
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Defne A Amado
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Michael Baer
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Andres Deik
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Lauren B Elman
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Michael Guo
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Ali G Hamedani
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - David J Irwin
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Aaron Lasker
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jennifer Orthmann-Murphy
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Colin C Quinn
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Thomas F Tropea
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Steven S Scherer
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Russell T Shinohara
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Roy H Hamilton
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Colin A Ellis
- From the Department of Neurology (A.B., J.C., M.A., L.D., K.J., R.A.P., D.A.A., M.B., A.D., L.B.E., M.G., A.G.H., D.J.I., A.L., J.O.-M., C.C.Q., T.F.T., S.S.S., R.H.H., C.A.E.), Penn Statistics in Imaging and Visualization Center (PennSIVE) (R.T.S.), Department of Biostatistics, Epidemiology, and Informatics, and Center for Biomedical Image Computing and Analytics (R.T.S.), Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
22
|
Hagiwara N, Harika N, Carmany EP, Shin Y, Eggly S, Jones SCT, Quillin J. Racial disparities in cancer genetic counseling encounters: study protocol for investigating patient-genetic counselor communication in the naturalistic clinical setting using a convergent mixed methods design. BMC Cancer 2023; 23:983. [PMID: 37845629 PMCID: PMC10578042 DOI: 10.1186/s12885-023-11486-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 10/06/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Despite decades of effort to reduce racial cancer disparities, Black people continue to die at higher rates from cancer than any other U.S. racial group. Because prevention is a key to the cost-effective and long-term control of cancer, the potential for cancer genetic counseling to play a central role in reducing racial cancer disparities is high. However, the benefits of genetic counseling are not equitable across race. Only 2% of genetic counselors self-identify as Black/African American, so most genetic counseling encounters with Black patients are racially discordant. Patients in racially discordant medical interactions tend to have poorer quality patient-provider communication and receive suboptimal clinical recommendations. One major factor that contributes to these healthcare disparities is racial bias. Drawing on findings from prior research, we hypothesize that genetic counselor providers' implicit racial prejudice will be associated negatively with the quality of patient-provider communication, while providers' explicit negative racial stereotypes will be associated negatively with the comprehensiveness of clinical discussions of cancer risk and genetic testing for Black (vs. White) patients. METHODS Using a convergent mixed methods research design, we will collect data from at least 15 genetic counseling providers, from two different institutions, and their 220 patients (approximately equal number of Black and White patients per provider) whose appointments are for a hereditary cancer condition. The data sources will include two provider surveys, two patient surveys, video- and/or audio-recordings of genetic counseling encounters, and medical chart reviews. The recorded cancer genetic counseling in-person and telehealth encounters will be analyzed both qualitatively and quantitatively to assess the quality of patient-provider communication and the comprehensiveness of clinical discussion. Those data will be linked to pre- and post-encounter survey data and data from medical chart reviews to test our hypotheses. DISCUSSION Findings from this multi-site study will highlight specific aspects of cancer genetic counseling encounters (patient-provider communication and clinical recommendations) that are directly associated with patient-centered outcomes (e.g., satisfaction, trust, genetic testing completion). Patient-provider communication and clinical recommendations are modifiable factors that can be integrated into current genetic counseling training curricula and thus can have immediate impact on genetic counseling training and practice.
Collapse
Affiliation(s)
- Nao Hagiwara
- Department of Public Health Sciences, University of Virginia, 200 Jeanette Lancaster Way, Charlottesville, VA, 22903, USA.
| | - Nadia Harika
- Department of Pediatrics, Virginia Commonwealth University, 1008 East Clay Street, B-011 Box 980270, Richmond, VA, 23219, USA
| | - Erin P Carmany
- Center for Molecular Medicine and Genetics, Wayne State University, 3127 Scott Hall, 540 E. Canfield Ave, Detroit, MI, 48201, USA
| | - Yongyun Shin
- Department of Biostatistics, Virginia Commonwealth University, 830 East Main Street, One Capitol Square 718, Richmond, VA, 23298, USA
| | - Susan Eggly
- Department of Oncology, Wayne State University, Karmanos Cancer Institute, 4100 John R, Detroit, MI, 48201, USA
| | - Shawn C T Jones
- Department of Psychology, Virginia Commonwealth University, 806 West Franklin Street, 23284, Richmond, VA, USA
| | - John Quillin
- Department of Pediatrics, Virginia Commonwealth University, 1008 East Clay Street, B-011 Box 980270, Richmond, VA, 23219, USA
| |
Collapse
|
23
|
Cole JJ, Sellitto AD, Baratta LR, Huecker JB, Balls-Berry JE, Gurnett CA. Social Determinants of Genetics Referral and Completion Rates Among Child Neurology Patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.12.23295450. [PMID: 37745339 PMCID: PMC10516043 DOI: 10.1101/2023.09.12.23295450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Objective To investigate clinical, social, and systems-level determinants predictive of genetics clinic referral and completion of genetics clinic visits among child neurology patients. Methods Electronic health record data were extracted from patients 0-18 years old who were evaluated in child neurology clinics at a single tertiary care institution between July 2018 to January 2020. Variables aligned with the Health Equity Implementation Framework. Referral and referral completion rates to genetics and cardiology clinics were compared among Black vs White patients using bivariate analysis. Demographic variables associated with genetics clinic referral and visit completion were identified using logistic regressions. Results In a cohort of 11,371 child neurology patients, 304 genetics clinic referrals and 82 cardiology clinic referrals were placed. In multivariate analysis of patients with Black or White ethnoracial identity (n=10,601), genetics clinic referral rates did not differ by race, but were significantly associated with younger age, rural address, neurodevelopmental disorder diagnosis, number of neurology clinic visits, and provider type. The only predictors of genetics clinic visit completion number of neurology clinic visits and race/ethnicity, with White patients being twice as likely as Black patients to complete the visit. Cardiology clinic referrals and visit completion did not differ by race/ethnicity. Interpretation Although race/ethnicity was not associated with differences in genetics clinic referral rates, White patients were twice as likely as Black patients to complete a genetics clinic visit after referral. Further work is needed to determine whether this is due to systemic/structural racism, differences in attitudes toward genetic testing, or other factors.
Collapse
Affiliation(s)
- Jordan J Cole
- Washington University in St. Louis, Department of Neurology
- University of Colorado, Department of Pediatrics
| | | | | | - Julia B Huecker
- Washington University in St. Louis, Center for Biostatistics & Data Science
| | | | | |
Collapse
|
24
|
Saylor KW, Klein WM, Calancie L, Lewis KL, Biesecker LG, Turbitt E, Roberts MC. Genetic Testing and Other Healthcare Use by Black and White Individuals in a Genomic Sequencing Study. Public Health Genomics 2023; 26:90-102. [PMID: 37544304 PMCID: PMC10614486 DOI: 10.1159/000533356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/27/2023] [Indexed: 08/08/2023] Open
Abstract
INTRODUCTION Early adopters play a critical role in the diffusion of medical innovations by spreading awareness, increasing acceptability, and driving demand. Understanding the role of race in the context of other characteristics of potential early adopters can shed light on disparities seen in the early implementation of genomic medicine. We aimed to understand the association between self-identified race and individual experience with genetic testing outside of the research context. METHODS We assessed factors associated with the odds of having ever received genetic testing prior to enrollment in a genomic sequencing study among 674 self-identified white and 407 self-identified African, African American, or Afro-Caribbean ("Black") individuals. RESULTS Controlling for individual determinants of healthcare use (demographics, personality traits, knowledge and attitudes, and health status), identifying as Black was associated with lower odds of prior genetic testing (OR = 0.43, 95% CI [0.27-0.68], p < 0.001). In contrast, self-identified race was not associated with the use of non-genetic clinical screening tests (e.g., echocardiogram, colonoscopy). Black and white individuals were similar on self-reported personality traits tied to early adoption but differed by sociodemographic and resource facilitators of early adoption. CONCLUSION Persistent racial disparities among early adopters may represent especially-entrenched disparities in access to and knowledge of genomic technologies in clinical settings.
Collapse
Affiliation(s)
- Katherine W. Saylor
- Department of Medical Ethics and Health Policy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - William M.P. Klein
- Behavioral Research Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Larissa Calancie
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Katie L. Lewis
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Leslie G. Biesecker
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Erin Turbitt
- Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia
| | - Megan C. Roberts
- Division of Pharmaceutical Outcomes and Policy, UNC Eshelman School of Pharmacy, Chapel Hill, NC, USA
| |
Collapse
|
25
|
Karpel H, Manderski E, Pothuri B. Frequency of actionable germline pathogenic variants identified through tumor next-generation sequencing in a gynecologic cancer cohort. Int J Gynecol Cancer 2023; 33:1044-1050. [PMID: 36889816 DOI: 10.1136/ijgc-2022-004142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023] Open
Abstract
BACKGROUND Tumor next-generation sequencing can identify potential germline pathogenic variants associated with cancer susceptibility. OBJECTIVE To describe the frequency of tumor sequencing results that met European Society of Medical Oncology (ESMO) recommendations for further germline genetic testing, and the frequency of germline variants among a cohort with gynecologic cancer. METHODS Patients with gynecologic cancer who underwent tumor sequencing between September 2019 and February 2022 in a large healthcare system in New York City were retrospectively identified. Eligible patients with suspected germline pathogenic variants on tumor sequencing were identified based on ESMO guidelines. Logistic regression was used to explore variables associated with referral and completion of germline testing. RESULTS Of 358 patients with gynecologic cancers who underwent tumor sequencing, 81 (22.6%) had ≥1 suspected germline variant according to ESMO guidelines. Of the 81 patients with qualifying tumor sequencing results, 56 (69.1%) received germline testing: 41/46 (89.1%) eligible patients with ovarian cancer and 15/33 (45.5%) with endometrial cancer. In the endometrial cancer cohort, 11/33 (33.3%) eligible patients were not referred for germline testing and the majority of these patients had tumor variants in genes commonly known to cause hereditary cancer. Of the 56 patients who underwent germline testing, 40 (71.4%) had pathogenic germline variants. In multivariable analysis, race/ethnicity other than non-Hispanic white was associated with lower odds of germline testing referral and completion (OR=0.1, 95% CI 0.01 to 0.5 and OR=0.2, 95% CI 0.04 to 0.6, respectively). CONCLUSION Given the high rate of pathogenic germline variant detection and the importance of identifying such variants for both patients and their family, it is imperative that eligible patients undergo germline testing. Additional education for providers on multidisciplinary guidelines and development of clinical pathways to ensure germline testing of suspected pathogenic variants identified on tumor sequencing is warranted, especially in light of the racial/ethnic inequity observed.
Collapse
Affiliation(s)
- Hannah Karpel
- New York University Grossman School of Medicine, New York, New York, USA
| | - Elizabeth Manderski
- Perlmutter Cancer Center, New York University Langone Health, New York, New York, USA
| | - Bhavana Pothuri
- Department of Obstetrics and Gynecology, Perlmutter Cancer Center, New York University Langone Health, New York, New York, USA
| |
Collapse
|
26
|
Matalon DR, Zepeda-Mendoza CJ, Aarabi M, Brown K, Fullerton SM, Kaur S, Quintero-Rivera F, Vatta M. Clinical, technical, and environmental biases influencing equitable access to clinical genetics/genomics testing: A points to consider statement of the American College of Medical Genetics and Genomics (ACMG). Genet Med 2023; 25:100812. [PMID: 37058144 DOI: 10.1016/j.gim.2023.100812] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 02/07/2023] [Indexed: 04/15/2023] Open
Affiliation(s)
- Dena R Matalon
- Division of Medical Genetics, Department of Pediatrics, Stanford Medicine, Stanford University, Stanford, CA
| | - Cinthya J Zepeda-Mendoza
- Divisions of Hematopathology and Laboratory Genetics and Genomics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Mahmoud Aarabi
- UPMC Medical Genetics and Genomics Laboratories, UPMC Magee-Womens Hospital, Pittsburgh, PA; Departments of Pathology and Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | | | - Stephanie M Fullerton
- Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, WA; Department of Bioethics & Humanities, University of Washington School of Medicine, Seattle, WA
| | - Shagun Kaur
- Department of Child Health, Phoenix Children's Hospital, University of Arizona College of Medicine-Phoenix, Phoenix, AZ
| | - Fabiola Quintero-Rivera
- Division of Genetic and Genomic Medicine, Departments of Pathology, Laboratory Medicine, and Pediatrics, University of California Irvine, Irvine, CA
| | | |
Collapse
|
27
|
Sei S, Ahadova A, Keskin DB, Bohaumilitzky L, Gebert J, von Knebel Doeberitz M, Lipkin SM, Kloor M. Lynch syndrome cancer vaccines: A roadmap for the development of precision immunoprevention strategies. Front Oncol 2023; 13:1147590. [PMID: 37035178 PMCID: PMC10073468 DOI: 10.3389/fonc.2023.1147590] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/09/2023] [Indexed: 04/11/2023] Open
Abstract
Hereditary cancer syndromes (HCS) account for 5~10% of all cancer diagnosis. Lynch syndrome (LS) is one of the most common HCS, caused by germline mutations in the DNA mismatch repair (MMR) genes. Even with prospective cancer surveillance, LS is associated with up to 50% lifetime risk of colorectal, endometrial, and other cancers. While significant progress has been made in the timely identification of germline pathogenic variant carriers and monitoring and early detection of precancerous lesions, cancer-risk reduction strategies are still centered around endoscopic or surgical removal of neoplastic lesions and susceptible organs. Safe and effective cancer prevention strategies are critically needed to improve the life quality and longevity of LS and other HCS carriers. The era of precision oncology driven by recent technological advances in tumor molecular profiling and a better understanding of genetic risk factors has transformed cancer prevention approaches for at-risk individuals, including LS carriers. MMR deficiency leads to the accumulation of insertion and deletion mutations in microsatellites (MS), which are particularly prone to DNA polymerase slippage during DNA replication. Mutations in coding MS give rise to frameshift peptides (FSP) that are recognized by the immune system as neoantigens. Due to clonal evolution, LS tumors share a set of recurrent and predictable FSP neoantigens in the same and in different LS patients. Cancer vaccines composed of commonly recurring FSP neoantigens selected through prediction algorithms have been clinically evaluated in LS carriers and proven safe and immunogenic. Preclinically analogous FSP vaccines have been shown to elicit FSP-directed immune responses and exert tumor-preventive efficacy in murine models of LS. While the immunopreventive efficacy of "off-the-shelf" vaccines consisting of commonly recurring FSP antigens is currently investigated in LS clinical trials, the feasibility and utility of personalized FSP vaccines with individual HLA-restricted epitopes are being explored for more precise targeting. Here, we discuss recent advances in precision cancer immunoprevention approaches, emerging enabling technologies, research gaps, and implementation barriers toward clinical translation of risk-tailored prevention strategies for LS carriers. We will also discuss the feasibility and practicality of next-generation cancer vaccines that are based on personalized immunogenic epitopes for precision cancer immunoprevention.
Collapse
Affiliation(s)
- Shizuko Sei
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Aysel Ahadova
- Department of Applied Tumor Biology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Derin B. Keskin
- Translational Immunogenomics Laboratory, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
- Broad Institute of The Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- Department of Computer Science, Metropolitan College, Boston University, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Lena Bohaumilitzky
- Department of Applied Tumor Biology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Johannes Gebert
- Department of Applied Tumor Biology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Magnus von Knebel Doeberitz
- Department of Applied Tumor Biology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Steven M. Lipkin
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Matthias Kloor
- Department of Applied Tumor Biology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Cooperation Unit Applied Tumor Biology, German Cancer Research Center Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| |
Collapse
|
28
|
A Canadian Provincial Screening Program for Lynch Syndrome. Am J Gastroenterol 2023; 118:345-353. [PMID: 36219179 DOI: 10.14309/ajg.0000000000002059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 10/06/2022] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Manitoba implemented the first Canadian provincial program of reflex screening through mismatch repair immunohistochemistry (MMR-IHC) for all colorectal cancers diagnosed at age 70 years or younger in December 2017. We evaluated compliance to universal reflex testing and for referrals to Genetics for individuals with MMR-deficient tumors. METHODS We searched the provincial pathology database with "adenocarcinoma" in the colorectal specimen pathology reports between March 2018 and December 2020. We cross-referenced with paper and electronic records in the Program of Genetics and Metabolism to determine whether patients with MMR-deficient tumors had been referred for Genetic assessment and what proportion of patients and first-degree relatives accepted an appointment and genetic testing. We performed logistic regression analysis to identify predictors of testing. RESULTS We identified 3,146 colorectal adenocarcinoma specimens (biopsies and surgical resections) from 1,692 unique individuals (mean age 68.66 years, male 57%). Of those aged 70 years or younger (n = 936), 89.4% received MMR-IHC screening. Individual pathologists (categorized by the highest, average, and lowest screening rates) were the biggest predictors of MMR-IHC screening on multivariable analysis (highest vs lowest: odds ratio 17.5, 95% confidence interval 6.05-50.67). While only 53.4% (n = 31) of 58 screen-positive cases were referred by pathologists for genetic assessment, other clinicians referred an additional 22.4% (n = 13), resulting in 75.8% overall referral rate of screen-positive cases. Thirteen (1.4%) patients (1.1%, aged 70 years or younger) were confirmed to experience Lynch syndrome through germline testing, and 8 first-degree relatives (an average of 1.6 per patient) underwent cascade genetic testing. DISCUSSION The first Canadian Lynch syndrome screening program has achieved high rates of reflex testing.
Collapse
|
29
|
Rodriguez NJ, Ricker C, Stoffel EM, Syngal S. Barriers and Facilitators to Genetic Education, Risk Assessment, and Testing: Considerations on Advancing Equitable Genetics Care. Gastroenterology 2023; 164:5-8. [PMID: 36529467 PMCID: PMC11009722 DOI: 10.1053/j.gastro.2022.11.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Nicolette Juliana Rodriguez
- Division of Gastroenterology, Brigham and Women's Hospital, Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Charité Ricker
- Norris Comprehensive Cancer Center, Division of Medical Oncology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Elena M Stoffel
- Division of Gastroenterology, Department of Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Sapna Syngal
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
30
|
Rodriguez NJ, Ricker C, Stoffel EM, Syngal S. Barriers and Facilitators to Genetic Education, Risk Assessment, and Testing: Considerations on Advancing Equitable Genetics Care. Clin Gastroenterol Hepatol 2023; 21:3-7. [PMID: 36549838 PMCID: PMC10609510 DOI: 10.1016/j.cgh.2022.10.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Nicolette Juliana Rodriguez
- Division of Gastroenterology, Brigham and Women's Hospital, Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Charité Ricker
- Norris Comprehensive Cancer Center, Division of Medical Oncology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Elena M Stoffel
- Division of Gastroenterology, Department of Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Sapna Syngal
- Division of Cancer Genetics and Prevention, Dana-Farber Cancer Institute, Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
31
|
del Carmen G, Reyes-Uribe L, Goyco D, Evans K, Bowen CM, Kinnison JL, Sepeda VO, Weber DM, Moskowitz J, Mork ME, Thirumurthi S, Lynch PM, Rodriguez-Bigas MA, Taggart MW, You YN, Vilar E. Colorectal surveillance outcomes from an institutional longitudinal cohort of lynch syndrome carriers. Front Oncol 2023; 13:1146825. [PMID: 37168379 PMCID: PMC10164917 DOI: 10.3389/fonc.2023.1146825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023] Open
Abstract
Objective Lynch Syndrome (LS) carriers have a significantly increased risk of developing colorectal cancer (CRC) during their lifetimes. Further stratification of this patient population may help in identifying additional risk factors that predispose to colorectal carcinogenesis. In most LS patients CRC may arise from adenomas, although an alternative non-polypoid carcinogenesis pathway has been proposed for PMS2 carriers. Using data from our institutional LS cohort, our aim was to describe our current colorectal screening outcomes with a focus on the incidence of adenomas in the context of different MMR genotypes and patient demographics such as gender, race, and ethnicity. Design We collected demographics, genetic, colonoscopy, and pathology results from a total of 163 LS carriers who obtained regular screening care at MD Anderson Cancer Center. Data were extracted from the electronic health records into a REDCap database for analysis. Logistic regressions were performed to measure the association between MMR variants and the likelihood of adenomas, advanced adenomas, and CRC. Then, we analyzed the cumulative incidences of these outcomes for the first 36 months following enrollment using Kaplan-Meier incidence curves, and Cox proportional hazard regressions. Results On multivariate analysis, age (≥45 years old) was associated with an increased risk of developing adenomas (P=0.034). Patients with a prior or active cancer status were less likely to develop adenomas (P=0.015), despite of the lack of association between surgical history with this outcome (P=0.868). We found no statistically significant difference in likelihood of adenoma development between MLH1 and MSH2/EPCAM, MSH6, and PMS2 carriers. Moreover, we observed no statistically significant difference in the likelihood of advanced adenomas or CRC for any measured covariates. On Cox proportional hazard, compared to MLH1 carriers, the incidence of adenomas was highest among MSH2/EPCAM carriers during for the first 36-months of follow-up (P<0.001). We observed a non-statistically significant trend for Hispanics having a higher and earlier cumulative incidence of adenomas compared to non-Hispanics (P=0.073). No MMR carrier was more likely to develop advanced adenomas. No difference in the incidence of CRC by MMR gene (P=0.198). Conclusion Screening recommendations for CRC in LS patients should be based on specific MMR variants and should also be tailored to consider patient demographics.
Collapse
Affiliation(s)
- Gabriel del Carmen
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- McGovern Medical School, University of Texas Health Science Center, Houston, TX, United States
| | - Laura Reyes-Uribe
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Daniel Goyco
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Kyera Evans
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Charles M. Bowen
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jennifer L. Kinnison
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Valerie O. Sepeda
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Diane M. Weber
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Julie Moskowitz
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maureen E. Mork
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Selvi Thirumurthi
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Patrick M. Lynch
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Miguel A. Rodriguez-Bigas
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Colorectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Melissa W. Taggart
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Y. Nancy You
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Colorectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: Eduardo Vilar,
| |
Collapse
|
32
|
Mighton C, Shickh S, Aguda V, Krishnapillai S, Adi-Wauran E, Bombard Y. From the patient to the population: Use of genomics for population screening. Front Genet 2022; 13:893832. [PMID: 36353115 PMCID: PMC9637971 DOI: 10.3389/fgene.2022.893832] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 09/26/2022] [Indexed: 10/22/2023] Open
Abstract
Genomic medicine is expanding from a focus on diagnosis at the patient level to prevention at the population level given the ongoing under-ascertainment of high-risk and actionable genetic conditions using current strategies, particularly hereditary breast and ovarian cancer (HBOC), Lynch Syndrome (LS) and familial hypercholesterolemia (FH). The availability of large-scale next-generation sequencing strategies and preventive options for these conditions makes it increasingly feasible to screen pre-symptomatic individuals through public health-based approaches, rather than restricting testing to high-risk groups. This raises anew, and with urgency, questions about the limits of screening as well as the moral authority and capacity to screen for genetic conditions at a population level. We aimed to answer some of these critical questions by using the WHO Wilson and Jungner criteria to guide a synthesis of current evidence on population genomic screening for HBOC, LS, and FH.
Collapse
Affiliation(s)
- Chloe Mighton
- Genomics Health Services Research Program, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Salma Shickh
- Genomics Health Services Research Program, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Vernie Aguda
- Genomics Health Services Research Program, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Centre for Medical Education, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Suvetha Krishnapillai
- Genomics Health Services Research Program, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Ella Adi-Wauran
- Genomics Health Services Research Program, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Yvonne Bombard
- Genomics Health Services Research Program, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
33
|
Bartley AN, Mills AM, Konnick E, Overman M, Ventura CB, Souter L, Colasacco C, Stadler ZK, Kerr S, Howitt BE, Hampel H, Adams SF, Johnson W, Magi-Galluzzi C, Sepulveda AR, Broaddus RR. Mismatch Repair and Microsatellite Instability Testing for Immune Checkpoint Inhibitor Therapy: Guideline From the College of American Pathologists in Collaboration With the Association for Molecular Pathology and Fight Colorectal Cancer. Arch Pathol Lab Med 2022; 146:1194-1210. [PMID: 35920830 DOI: 10.5858/arpa.2021-0632-cp] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2022] [Indexed: 11/06/2022]
Abstract
CONTEXT.— The US Food and Drug Administration (FDA) approved immune checkpoint inhibitor therapy for patients with advanced solid tumors that have DNA mismatch repair defects or high levels of microsatellite instability; however, the FDA provided no guidance on which specific clinical assays should be used to determine mismatch repair status. OBJECTIVE.— To develop an evidence-based guideline to identify the optimal clinical laboratory test to identify defects in DNA mismatch repair in patients with solid tumor malignancies who are being considered for immune checkpoint inhibitor therapy. DESIGN.— The College of American Pathologists convened an expert panel to perform a systematic review of the literature and develop recommendations. Using the National Academy of Medicine-endorsed Grading of Recommendations Assessment, Development and Evaluation approach, the recommendations were derived from available evidence, strength of that evidence, open comment feedback, and expert panel consensus. Mismatch repair immunohistochemistry, microsatellite instability derived from both polymerase chain reaction and next-generation sequencing, and tumor mutation burden derived from large panel next-generation sequencing were within scope. RESULTS.— Six recommendations and 3 good practice statements were developed. More evidence and evidence of higher quality were identified for colorectal cancer and other cancers of the gastrointestinal (GI) tract than for cancers arising outside the GI tract. CONCLUSIONS.— An optimal assay depends on cancer type. For most cancer types outside of the GI tract and the endometrium, there was insufficient published evidence to recommend a specific clinical assay. Absent published evidence, immunohistochemistry is an acceptable approach readily available in most clinical laboratories.
Collapse
Affiliation(s)
- Angela N Bartley
- From the Department of Pathology, St. Joseph Mercy Hospital, Ann Arbor, Michigan (Bartley)
| | - Anne M Mills
- From the Department of Pathology, University of Virginia, Charlottesville (Mills)
| | - Eric Konnick
- From the Department of Laboratory Medicine and Pathology, University of Washington, Seattle (Konnick)
| | - Michael Overman
- From the Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston (Overman)
| | - Christina B Ventura
- From Surveys, College of American Pathologists, Northfield, Illinois (Ventura, Colasacco)
| | - Lesley Souter
- From Methodology Consultant, Smithville, Ontario, Canada (Souter)
| | - Carol Colasacco
- From Surveys, College of American Pathologists, Northfield, Illinois (Ventura, Colasacco)
| | - Zsofia K Stadler
- From the Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York (Stadler)
| | - Sarah Kerr
- From Hospital Pathology Associates, PA, Minneapolis, Minnesota (Kerr)
| | - Brooke E Howitt
- From the Department of Pathology, Stanford University, Stanford, California (Howitt)
| | - Heather Hampel
- From the Department of Internal Medicine, The Ohio State University, Columbus (Hampel)
| | - Sarah F Adams
- From the Department of Obstetrics & Gynecology, University of New Mexico, Albuquerque (Adams)
| | - Wenora Johnson
- From Fight Colorectal Cancer, Springfield, Missouri (Johnson)
| | - Cristina Magi-Galluzzi
- From the Department of Pathology, University of Alabama at Birmingham, Birmingham (Magi-Galluzzi)
| | - Antonia R Sepulveda
- From the Department of Pathology, George Washington University, Washington, District of Columbia (Sepulveda)
| | - Russell R Broaddus
- From the Department of Pathology & Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill (Broaddus)
| |
Collapse
|
34
|
Singh V, Mezzacappa C, Gershkovich P, Di Giovanna J, Ganzak A, Gibson J, Sinard J, Xicola RM, Llor X. Systems approach to enhance Lynch syndrome diagnosis through tumour testing. J Med Genet 2022; 60:533-539. [PMID: 36115663 PMCID: PMC10020126 DOI: 10.1136/jmg-2022-108770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/08/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Guidelines recommend universal mismatch repair (MMR) tumour testing of colorectal adenocarcinomas (CRCs) to screen for Lynch syndrome (LS). However, its implementation remains disjointed and referral for genetic testing dismal, particularly among minorities. We aimed to increase referral, cancer genetic testing and eventually LS diagnosis by developing the CLEAR LS (Closed Loop Enhanced Assessment and Referral for Lynch Syndrome) intervention, a systems approach which in the second phase was automated. METHODS This is a cohort study of all patients diagnosed with CRC at an academic centre between 1 January 2012, when implementation of universal CRC testing began, and 31 January 2021. The original cohort spanned through 31 May 2015. Tumour testing included MMR immunohistochemistry, followed by BRAF V600E/MLH1 promoter methylation testing when indicated. The intervention included a manual phase (1 June 2015 through 31 July 2018), which systematised pathology screening and cancer genetics (CG) referral mechanisms, and an automated phase (1 August 2018 through 31 January 2021) using computer programming. RESULTS A total of 249/1541 CRC (17.38%) had MMR loss of expression and 129 (8.37%) qualified for CG evaluation. Referral was 27.58% in the original cohort and 92.1% in the intervention (p<0.001). Patients seen by CG among referred were 27.58% in the original cohort and 74.3% in the intervention (p two-sided<0.001). The distribution of race/ethnicity among patients qualifying and referred for CG evaluation was not significantly different across cohorts. LS diagnosis increased from 0.56% (original cohort) to 1.43% (intervention). Cost per new diagnosis of LS decreased from US$173 675 to $87 960 from original cohort to intervention. CONCLUSION Implementation of systematic case identification and referral support mechanisms significantly increased the proportion of patients undergoing genetic testing and doubled the percentage of patients diagnosed with LS with no referral differences across racial/ethnic groups.
Collapse
Affiliation(s)
- Vinit Singh
- Department of Medicine, Yale University, New Haven, Connecticut, USA
| | | | - Peter Gershkovich
- Department of Pathology, Yale University, New Haven, Connecticut, USA
| | - Jessica Di Giovanna
- Cancer Genetics and Prevention Program, Yale-New Haven Hospital, New Haven, Connecticut, USA
| | - Amanda Ganzak
- Cancer Genetics and Prevention Program, Yale-New Haven Hospital, New Haven, Connecticut, USA
| | - Joanna Gibson
- Department of Pathology, Yale University, New Haven, Connecticut, USA
| | - John Sinard
- Department of Pathology, Yale University, New Haven, Connecticut, USA
| | - Rosa M Xicola
- Department of Medicine, Yale University, New Haven, Connecticut, USA
| | - Xavier Llor
- Department of Medicine, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
35
|
Dellefave-Castillo LM, Cirino AL, Callis TE, Esplin ED, Garcia J, Hatchell KE, Johnson B, Morales A, Regalado E, Rojahn S, Vatta M, Nussbaum RL, McNally EM. Assessment of the Diagnostic Yield of Combined Cardiomyopathy and Arrhythmia Genetic Testing. JAMA Cardiol 2022; 7:966-974. [PMID: 35947370 PMCID: PMC9366660 DOI: 10.1001/jamacardio.2022.2455] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Importance Genetic testing can guide management of both cardiomyopathies and arrhythmias, but cost, yield, and uncertain results can be barriers to its use. It is unknown whether combined disease testing can improve diagnostic yield and clinical utility for patients with a suspected genetic cardiomyopathy or arrhythmia. Objective To evaluate the diagnostic yield and clinical management implications of combined cardiomyopathy and arrhythmia genetic testing through a no-charge, sponsored program for patients with a suspected genetic cardiomyopathy or arrhythmia. Design, Setting, and Participants This cohort study involved a retrospective review of DNA sequencing results for cardiomyopathy- and arrhythmia-associated genes. The study included 4782 patients with a suspected genetic cardiomyopathy or arrhythmia who were referred for genetic testing by 1203 clinicians; all patients participated in a no-charge, sponsored genetic testing program for cases of suspected genetic cardiomyopathy and arrhythmia at a single testing site from July 12, 2019, through July 9, 2020. Main Outcomes and Measures Positive gene findings from combined cardiomyopathy and arrhythmia testing were compared with findings from smaller subtype-specific gene panels and clinician-provided diagnoses. Results Among 4782 patients (mean [SD] age, 40.5 [21.3] years; 2551 male [53.3%]) who received genetic testing, 39 patients (0.8%) were Ashkenazi Jewish, 113 (2.4%) were Asian, 571 (11.9%) were Black or African American, 375 (7.8%) were Hispanic, 2866 (59.9%) were White, 240 (5.0%) were of multiple races and/or ethnicities, 138 (2.9%) were of other races and/or ethnicities, and 440 (9.2%) were of unknown race and/or ethnicity. A positive result (molecular diagnosis) was confirmed in 954 of 4782 patients (19.9%). Of those, 630 patients with positive results (66.0%) had the potential to inform clinical management associated with adverse clinical outcomes, increased arrhythmia risk, or targeted therapies. Combined cardiomyopathy and arrhythmia gene panel testing identified clinically relevant variants for 1 in 5 patients suspected of having a genetic cardiomyopathy or arrhythmia. If only patients with a high suspicion of genetic cardiomyopathy or arrhythmia had been tested, at least 137 positive results (14.4%) would have been missed. If testing had been restricted to panels associated with the clinician-provided diagnostic indications, 75 of 689 positive results (10.9%) would have been missed; 27 of 75 findings (36.0%) gained through combined testing involved a cardiomyopathy indication with an arrhythmia genetic finding or vice versa. Cascade testing of family members yielded 402 of 958 positive results (42.0%). Overall, 2446 of 4782 patients (51.2%) had only variants of uncertain significance. Patients referred for arrhythmogenic cardiomyopathy had the lowest rate of variants of uncertain significance (81 of 176 patients [46.0%]), and patients referred for catecholaminergic polymorphic ventricular tachycardia had the highest rate (48 of 76 patients [63.2%]). Conclusions and Relevance In this study, comprehensive genetic testing for cardiomyopathies and arrhythmias revealed diagnoses that would have been missed by disease-specific testing. In addition, comprehensive testing provided diagnostic and prognostic information that could have potentially changed management and monitoring strategies for patients and their family members. These results suggest that this improved diagnostic yield may outweigh the burden of uncertain results.
Collapse
Affiliation(s)
- Lisa M Dellefave-Castillo
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Allison L Cirino
- Cardiovascular Division, Brigham and Women's Hospital, Boston, Massachusetts.,Institute of Health Professions, Massachusetts General Hospital, Boston
| | | | | | - John Garcia
- Invitae Corporation, San Francisco, California
| | | | | | - Ana Morales
- Invitae Corporation, San Francisco, California
| | | | | | | | | | - Elizabeth M McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
36
|
Esplin ED, Nielsen SM, Bristow SL, Garber JE, Hampel H, Rana HQ, Samadder NJ, Shore ND, Nussbaum RL. Universal Germline Genetic Testing for Hereditary Cancer Syndromes in Patients With Solid Tumor Cancer. JCO Precis Oncol 2022; 6:e2100516. [PMID: 36108258 PMCID: PMC9489188 DOI: 10.1200/po.21.00516] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Affiliation(s)
| | | | | | | | - Heather Hampel
- Division of Clinical Cancer Genomics, Department of Medical Oncology & Therapeutic Research, City of Hope National Cancer Center, Duarte, CA
| | | | - N Jewel Samadder
- Center for Individualized Medicine, Mayo Clinic, Phoenix, AZ.,Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Phoenix, AZ.,Department of Clinical Genomics, Mayo Clinic, Phoenix, AZ
| | - Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC
| | | |
Collapse
|
37
|
Brown TJ, Massa RC. Challenges in the Management of Patients With HER2-Amplified Colorectal Cancer. JCO Oncol Pract 2022; 18:555-556. [PMID: 35786957 PMCID: PMC9377689 DOI: 10.1200/op.22.00361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/09/2022] [Indexed: 08/03/2023] Open
Affiliation(s)
- Timothy J. Brown
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
- Penn Center for Cancer Care Innovation, University of Pennsylvania, Philadelphia, PA
| | - Ryan C. Massa
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
38
|
Khoury MJ, Bowen S, Dotson WD, Drzymalla E, Green RF, Goldstein R, Kolor K, Liburd LC, Sperling LS, Bunnell R. Health equity in the implementation of genomics and precision medicine: A public health imperative. Genet Med 2022; 24:1630-1639. [PMID: 35482015 PMCID: PMC9378460 DOI: 10.1016/j.gim.2022.04.009] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/05/2022] [Accepted: 04/05/2022] [Indexed: 12/24/2022] Open
Abstract
Recent reviews have emphasized the need for a health equity agenda in genomics research. To ensure that genomic discoveries can lead to improved health outcomes for all segments of the population, a health equity agenda needs to go beyond research studies. Advances in genomics and precision medicine have led to an increasing number of evidence-based applications that can reduce morbidity and mortality for millions of people (tier 1). Studies have shown lower implementation rates for selected diseases with tier 1 applications (familial hypercholesterolemia, Lynch syndrome, hereditary breast and ovarian cancer) among racial and ethnic minority groups, rural communities, uninsured or underinsured people, and those with lower education and income. We make the case that a public health agenda is needed to address disparities in implementation of genomics and precision medicine. Public health actions can be centered on population-specific needs and outcomes assessment, policy and evidence development, and assurance of delivery of effective and ethical interventions. Crucial public health activities also include engaging communities, building coalitions, improving genetic health literacy, and building a diverse workforce. Without concerted public health action, further advances in genomics with potentially broad applications could lead to further widening of health disparities in the next decade.
Collapse
Affiliation(s)
- Muin J Khoury
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA.
| | - Scott Bowen
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA
| | - W David Dotson
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA
| | - Emily Drzymalla
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA
| | - Ridgely F Green
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA
| | - Robert Goldstein
- Office of the Associate Director of Policy and Strategy, Centers for Disease Control and Prevention, Atlanta, GA; Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Katherine Kolor
- Office of Genomics and Precision Public Health, Office of Science, Centers for Disease Control and Prevention, Atlanta, GA
| | - Leandris C Liburd
- Office of Minority Health and Health Equity, Centers for Disease Control and Prevention, Atlanta, GA
| | | | - Rebecca Bunnell
- Office of Science, Centers for Disease Control and Prevention, Atlanta, GA
| |
Collapse
|
39
|
Modell SM, Schlager L, Allen CG, Marcus G. Medicaid Expansions: Probing Medicaid's Filling of the Cancer Genetic Testing and Screening Space. Healthcare (Basel) 2022; 10:1066. [PMID: 35742117 PMCID: PMC9223044 DOI: 10.3390/healthcare10061066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/25/2022] [Accepted: 06/05/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer is the third largest source of spending for Medicaid in the United States. A working group of the American Public Health Association Genomics Forum Policy Committee reviewed 133/149 pieces of literature addressing the impact of Medicaid expansion on cancer screening and genetic testing in underserved groups and the general population. Breast and colorectal cancer screening rates improved during very early Medicaid expansion but displayed mixed improvement thereafter. Breast cancer screening rates have remained steady for Latina Medicaid enrollees; colorectal cancer screening rates have improved for African Americans. Urban areas have benefited more than rural. State programs increasingly cover BRCA1/2 and Lynch syndrome genetic testing, though testing remains underutilized in racial and ethnic groups. While increased federal matching could incentivize more states to engage in Medicaid expansion, steps need to be taken to ensure that they have an adequate distribution of resources to increase screening and testing utilization.
Collapse
Affiliation(s)
- Stephen M. Modell
- Epidemiology, Center for Public Health and Community Genomics, School of Public Health, University of Michigan, M5409 SPH II, 1415 Washington Heights, Ann Arbor, MI 48109, USA
| | - Lisa Schlager
- Public Policy, FORCE: Facing Our Risk of Cancer Empowered, 16057 Tampa Palms Boulevard W, PMB #373, Tampa, FL 33647, USA;
| | - Caitlin G. Allen
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, 22 Westedge, Room 213, Charleston, SC 29403, USA;
| | - Gail Marcus
- Genetics and Newborn Screening Unit, North Carolina Department of Health and Human Services, C/O CDSA of the Cape Fear, 3311 Burnt Mill Drive, Wilmington, NC 28403, USA;
| |
Collapse
|
40
|
Hampel H, Yurgelun MB. Point/Counterpoint: Is It Time for Universal Germline Genetic Testing for all GI Cancers? J Clin Oncol 2022; 40:2681-2692. [PMID: 35649230 DOI: 10.1200/jco.21.02764] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Use of germline genetic testing among patients with cancer is increasing because of (1) the availability of multigene panel tests that include multiple cancer susceptibility genes in a single test, (2) decreased costs of these tests and improvements in insurance coverage, and (3) US Food and Drug Administration-approval of genotype-directed therapies such as poly(ADP-ribose) polymerase inhibitors for individuals with certain cancers and pathogenic germline variants in BRCA1 and BRCA2 (with possible benefits with other genes in the homologous repair deficiency pathway). In addition, National Comprehensive Cancer Network guidelines have already endorsed germline genetic testing for all patients with certain cancer types (epithelial ovarian cancer, exocrine pancreatic cancer, and high-grade/metastatic prostate cancer), regardless of age or personal/family history of cancer. Herein, we debate the pros and cons of offering germline multigene panel testing to all patients diagnosed with any GI cancer. The authors agree that it may just be a matter of time before germline multigene panel testing is offered to all patients with cancer; however, this article will highlight some of the benefits, risks, and limitations of this approach so that research can help fill some of the gaps to ensure that genetic medicine continues to be implemented in ways that improve real-world patient care and outcomes.
Collapse
Affiliation(s)
- Heather Hampel
- Division of Clinical Cancer Genomics, City of Hope National Medical Center, Duarte, CA
| | - Matthew B Yurgelun
- Dana-Farber Cancer Institute, Brigham & Women's Hospital, and Harvard Medical School, Boston, MA
| |
Collapse
|
41
|
Abstract
Contrary to decreasing incidence rate of colorectal cancer (CRC) in older adults, incidence rates have nearly doubled in younger adults (age <50 years) in the United States since the early 1990s. A similar increase has been observed across the globe. Despite overall population trends in aging, about 15% of CRCs will be diagnosed in younger adults by 2030. The mechanisms and factors contributing to early-onset CRC (EOCRC) remain puzzling, especially because most young adults diagnosed with CRC have no known risk factors or predisposing conditions, such as family history of CRC or polyps or a hereditary syndrome (eg, Lynch syndrome, polyposis). In this up-to-date review, we discuss the current knowledge of EOCRC, including epidemiology, risk factors, clinical and molecular features, treatment and survival, and recognition and screening strategies.
Collapse
Affiliation(s)
- Pooja Dharwadkar
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, Zuckerberg San Francisco General, Building 5, 3rd Floor, Suite 3D, 1001 Potrero Avenue, San Francisco, CA 94110, USA
| | - Timothy A Zaki
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Caitlin C Murphy
- UTHealth School of Public Health, Suite 2618, 7000 Fannin Street, Houston, TX 77030, USA.
| |
Collapse
|
42
|
Cost-effectiveness of population-wide genomic screening for Lynch syndrome in the United States. Genet Med 2022; 24:1017-1026. [PMID: 35227606 DOI: 10.1016/j.gim.2022.01.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Genomic screening for Lynch syndrome (LS) could prevent colorectal cancer (CRC) by identifying high-risk patients and instituting intensive CRC screening. We estimated the cost-effectiveness of a population-wide LS genomic screening vs family history-based screening alone in an unselected US population. METHODS We developed a decision-analytic Markov model including health states for precancer, stage-specific CRC, and death and assumed an inexpensive test cost of $200. We conducted sensitivity and threshold analyses to evaluate model uncertainty. RESULTS Screening unselected 30-year-olds for LS variants resulted in 48 (95% credible range [CR] = 35-63) fewer overall CRC cases per 100,000 screened individuals, leading to 187 quality-adjusted life-years (QALYs; 95% CR = 123-260) gained at an incremental cost of $24.6 million (95% CR = $20.3 million-$29.1 million). The incremental cost-effectiveness ratio was $132,200, with an 8% and 71% probability of being cost-effective at $100,000 and $150,000 per QALY willingness-to-pay thresholds, respectively. CONCLUSION Population LS screening may be cost-effective in younger patient populations under a $150,000 willingness-to-pay per QALY threshold and with a relatively inexpensive test cost. Further reductions in testing costs and/or the inclusion of LS testing within a broader multiplex screening panel are needed for screening to become highly cost-effective.
Collapse
|
43
|
Einarsson H, Runarsdottir JR, Tryggvason T, Snaebjornsson P, Smaradottir A, Stefansdottir V, Thoroddsen A, Arngrimsson R, Jonasson JG, Haraldsdottir S. Universal tumor screening in a population with MSH6- and PMS2-associated Lynch syndrome. Genet Med 2022; 24:999-1007. [PMID: 35172941 DOI: 10.1016/j.gim.2022.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 10/19/2022] Open
Abstract
PURPOSE Universal screening for Lynch syndrome (LS) on resected colorectal carcinomas (CRCs) and endometrial carcinomas (ECs) was implemented in Iceland in 2017 using immunohistochemistry (IHC) for mismatch repair (MMR) proteins. We examined the efficacy of the universal screening algorithm to detect LS and the diagnostic accuracy of MMR IHC by comparing results with a population-based genotype database. METHODS All patients diagnosed with CRC or EC per the Icelandic Cancer Registry from 2017 to 2019 who had tumor MMR IHC performed were included. Pathology reports and patient charts were reviewed. MMR IHC stains were crossmatched with genotyping results obtained from the deCODE database. RESULTS IHC staining was done on 404 patients with CRC and 74 patients with EC. A total of 61 (15.1%) patients with CRC and 15 (20.3%) patients with EC were MMR-deficient. MMR IHC had 88.9% sensitivity in identifying patients with LS and a positive predictive value of 10.7%. Only 50% of individuals were appropriately referred for genetic testing, leading to underdiagnosis of LS. CONCLUSION Universal screening for LS using MMR protein IHC in CRC and EC accurately identified patients appropriate for genetic testing in a population with MSH6 and PMS2 LS predominance. Because of lack of referral to genetic counseling, only 50% of patients with LS were identified through the screening algorithm.
Collapse
Affiliation(s)
- Haukur Einarsson
- Department of Pathology, Landspitali University Hospital of Iceland, Reykjavik, Iceland
| | | | - Thordur Tryggvason
- Department of Pathology, Landspitali University Hospital of Iceland, Reykjavik, Iceland
| | - Petur Snaebjornsson
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Agnes Smaradottir
- Department of Oncology, Landspitali University Hospital of Iceland, Reykjavik, Iceland
| | - Vigdis Stefansdottir
- Department of Genetics and Molecular Medicine, Landspitali University Hospital of Iceland, Reykjavik, Iceland
| | - Asgeir Thoroddsen
- Department of Obstetrics and Gynecology, Landspitali University Hospital of Iceland, Reykjavik, Iceland
| | - Reynir Arngrimsson
- Department of Genetics and Molecular Medicine, Landspitali University Hospital of Iceland, Reykjavik, Iceland; Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Jon Gunnlaugur Jonasson
- Department of Pathology, Landspitali University Hospital of Iceland, Reykjavik, Iceland; Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Sigurdis Haraldsdottir
- Department of Oncology, Landspitali University Hospital of Iceland, Reykjavik, Iceland; Faculty of Medicine, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
44
|
Crain PR, Zepp JM, Gille S, Jenkins L, Kauffman TL, Shuster E, Goddard KAB, Wilfond BS, Hunter JE. Identifying patients with Lynch syndrome using a universal tumor screening program in an integrated healthcare system. Hered Cancer Clin Pract 2022; 20:17. [PMID: 35436948 PMCID: PMC9014602 DOI: 10.1186/s13053-022-00217-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 03/01/2022] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION Lynch syndrome (LS) is associated with an increased risk of colorectal (CRC) and endometrial (EC) cancers. Universal tumor screening (UTS) of all individuals diagnosed with CRC and EC is recommended to increase identification of LS. Kaiser Permanente Northwest (KPNW) implemented a UTS program for LS among individuals newly diagnosed with CRC in January 2016 and EC in November 2016. UTS at KPNW begins with immunohistochemistry (IHC) of tumor tissue to determine loss of mismatch repair proteins associated with LS (MLH1, MSH2, MSH6, and PMS2)., IHC showing loss of MLH1 is followed by reflex testing (automatic testing) to detect the presence of the BRAF V600E variant (in cases of CRC) and MLH1 promoter hypermethylation to rule out likely sporadic cases. MATERIALS AND METHODS Individuals newly diagnosed with CRC and EC were identified between the initiation of the respective UTS programs and July 2018. Electronic medical records were reviewed to extract patient data related to UTS, including IHC and reflex testing results, date of referrals to the genetics department, and results of germline genetic testing for LS. RESULTS 313 out of 362 individuals diagnosed with CRC and 61 out of 64 individuals diagnosed with EC who were eligible were screened by IHC for LS. Most (47/52 or 90%, including 46/49 CRC and 1/3 EC) individuals that were not screened by IHC only had a biopsy sample available. Fourteen individuals (3.7% overall, including 13/313 CRC and 1/61 EC) received an abnormal result after reflex testing and were referred for genetic counseling. Of these, 10 individuals (71% overall, including 9/13 CRC and 1/1 EC) underwent germline genetic testing for LS. Five individuals diagnosed with CRC were found to have pathogenic variants. in PMS2 (n = 3), MLH1 (n = 1), and MSH6 (n = 1). No pathogenic variants were identified in individuals diagnosed with EC. CONCLUSIONS UTS identified individuals at risk for LS. Most individuals who screened positive for LS had follow-up germline genetic testing for LS. The consistent use of biopsy samples is an opportunity to improve UTS.
Collapse
Affiliation(s)
- Philip R Crain
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Jamilyn M Zepp
- Department of Translational and Applied Genomics, Center for Health Research, Kaiser Permanente Northwest, Portland, OR, USA
| | - Sara Gille
- Center for Health Research, Kaiser Permanente Northwest, Portland, OR, USA
| | - Lindsay Jenkins
- Center for Health Research, Kaiser Permanente Northwest, Portland, OR, USA
| | - Tia L Kauffman
- Department of Translational and Applied Genomics, Center for Health Research, Kaiser Permanente Northwest, Portland, OR, USA
| | - Elizabeth Shuster
- Center for Health Research, Kaiser Permanente Northwest, Portland, OR, USA
| | - Katrina A B Goddard
- Department of Translational and Applied Genomics, Center for Health Research, Kaiser Permanente Northwest, Portland, OR, USA
| | - Benjamin S Wilfond
- Treuman Katz Center for Pediatric Bioethics, Department of Pediatrics, Seattle Children's Research Institute and Hospital, University of Washington School of Medicine, Seattle, WA, USA
| | - Jessica Ezzell Hunter
- Department of Translational and Applied Genomics, Center for Health Research, Kaiser Permanente Northwest, Portland, OR, USA.
| |
Collapse
|
45
|
Rutter CM, May FP, Coronado GD, Pujol TA, Thomas EG, Cabreros I. Racism Is a Modifiable Risk Factor: Relationships Among Race, Ethnicity, and Colorectal Cancer Outcomes. Gastroenterology 2022; 162:1053-1055. [PMID: 34942173 DOI: 10.1053/j.gastro.2021.12.251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 01/16/2023]
Affiliation(s)
| | - Folasade P May
- David Geffen School of Medicine and UCLA Kaiser Permanente Center for Health Equity, University of California, Los Angeles, California
| | | | | | | | | |
Collapse
|
46
|
Douglas MP, Kumar A. Analyzing Precision Medicine Utilization with Real-World Data: A Scoping Review. J Pers Med 2022; 12:557. [PMID: 35455673 PMCID: PMC9025578 DOI: 10.3390/jpm12040557] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022] Open
Abstract
Precision medicine (PM), specifically genetic-based testing, is currently used in over 140,000 individual tests to inform the clinical management of disease. Though several databases (e.g., the NIH Genetic Testing Registry) demonstrate the availability of these sequencing-based tests, we do not currently understand the extent to which these tests are used. There exists a need to synthesize the body of real-world data (RWD) describing the use of sequencing-based tests to inform their appropriate use. To accomplish this, we performed a scoping review to examine what RWD sources have been used in studies of PM utilization between January 2015 and August 2021 to characterize the use of genome sequencing (GS), exome sequencing (ES), tumor sequencing (TS), next-generation sequencing-based panels (NGS), gene expression profiling (GEP), and pharmacogenomics (PGx) panels. We abstracted variables describing the use of these types of tests and performed a descriptive statistical analysis. We identified 440 articles in our search and included 72 articles in our study. Publications based on registry databases were the most common, followed by studies based on private insurer administrative claims. Slightly more than one-third (38%) used integrated datasets. Two thirds (67%) of the studies focused on the use of tests for oncological clinical applications. We summarize the RWD sources used in peer-reviewed literature on the use of PM. Our findings will help improve future study design by encouraging the use of centralized databases and registries to track the implementation and use of PM.
Collapse
Affiliation(s)
- Michael P. Douglas
- Center for Translational and Policy Research on Precision Medicine (TRANSPERS), Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Anika Kumar
- School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA;
| |
Collapse
|
47
|
Wehbe A, Manning M, Assad H, Purrington KS, Simon MS. Uptake of genetic counseling and testing in a clinic-based population of women with breast cancer. Cancer Med 2022; 11:3304-3311. [PMID: 35322585 PMCID: PMC9468430 DOI: 10.1002/cam4.4684] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/09/2021] [Accepted: 01/06/2022] [Indexed: 11/06/2022] Open
Abstract
Background The study was conducted to evaluate racial differences in referral and uptake of genetic counseling (GC) in a clinic‐based population of women with breast cancer. Methods Medical records of 150 breast cancer patients at the Karmanos Cancer Institute were reviewed to determine eligibility for GC according to National Comprehensive Cancer Network guidelines, GC referral rates, and appointment completion rates. Logistic regression was used to assess the relationship between demographic and clinical factors and GC eligibility and referral. Results The mean age at diagnosis was 57.1 (SD 12.6) and 66% of the women were Black. There were 91 women (60.7%) eligible for GC and of those, 54 (61.4%) were referred. After multivariable analyses, factors associated with reduced eligibility were older age at diagnosis (OR = 0.91, 95% CI [0.87,0.95]) and Black race (OR = 0.37, 95% CI [0.15, 0.96]). After additional multivariable analysis, eligibility was associated with an increased likelihood of referral (OR = 5.97, 95% CI [2.29, 15.56]), however, Medicare versus private insurance was associated with a lower likelihood for referral (OR = 0.32, 95% CI [0.12–0.80]. Of those referred, 49 (76.6%) completed an appointment, and 47 had genetic testing. Women with Medicare were also less likely to complete an appointment. Race had no impact on referral or appointment completion. Conclusions There were no racial differences in GC referral or appointment completion in a clinic‐based sample of women with breast cancer. Further interventions are needed to promote increased referral and appointment completion for women with breast cancer who are eligible for GC.
Collapse
Affiliation(s)
- Alexandra Wehbe
- Wayne State University School of Medicine, Detroit, Michigan, USA.,Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan, USA
| | - Mark Manning
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan, USA.,Department of Psychology, Oakland University, Rochester Hills, Michigan, USA
| | - Hadeel Assad
- Wayne State University School of Medicine, Detroit, Michigan, USA.,Department of Oncology, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan, USA
| | - Kristen S Purrington
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan, USA
| | - Michael S Simon
- Population Studies and Disparities Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan, USA.,Department of Oncology, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan, USA
| |
Collapse
|
48
|
Patient navigation for hereditary colorectal cancer: Design of a randomized controlled trial to determine the effectiveness of pathways to genetic counseling. Contemp Clin Trials 2022; 116:106735. [PMID: 35331945 DOI: 10.1016/j.cct.2022.106735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/28/2022] [Accepted: 03/17/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Diagnosis of Lynch and other hereditary colorectal cancer (CRC) syndromes through germline genetic testing has important implications for treatment and risk-management, yet guideline-recommended genetic counseling referral and attendance is suboptimal. METHODS Our team developed an adapted patient navigation program-Pathways to Genetic Counseling-to address multilevel barriers to genetic counseling referral and receipt. This paper describes the methods of a randomized controlled trial (RCT) testing Pathways to Genetic Counseling's effectiveness at increasing genetic counseling attendance in the University of Washington Medicine health system. We will identify CRC patients eligible for genetic counseling (diagnosed before age 50 or at any age with evidence of inherited mismatch repair deficiency) through a combination of structured electronic health record queries and manual chart review. Patients will be randomized 1:1 prior to consent and receive either care as usual (no contact) or be invited to participate in patient navigation. We will use chart review to compare rates of genetic counseling referral and attendance within six months of randomization, regardless of patients' engagement with navigation. We plan to identify and randomize 161 eligible CRC patients over a nine-month period beginning in late 2021. DISCUSSION Our pragmatic RCT design will provide real-world data on the potential for patient navigation to address longstanding care gaps in preventive genomic medicine. If effective, we hope to pilot Pathways to Genetic Counseling in additional settings with a long-term goal of improving appropriate diagnosis of hereditary CRC syndromes and subsequent cascade screening of eligible family members.
Collapse
|
49
|
Khan A, Rogers CR, Kennedy CD, Lopez A, Jeter J. Genetic Evaluation for Hereditary Cancer Syndromes Among African Americans: A Critical Review. Oncologist 2022; 27:285-291. [PMID: 35380723 PMCID: PMC8982373 DOI: 10.1093/oncolo/oyab082] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/24/2021] [Indexed: 11/12/2022] Open
Abstract
Abstract
While hereditary cancer syndromes have been described and studied for centuries, the completion of the human genome project fueled accelerated progress in precision medicine due to the introduction of genetic testing in the 1990s, creating avenues for tailored treatments and medical management options. However, genetic testing has not benefited everyone equitably, with nearly all of the published work based on individuals of non-Hispanic White/European ancestry. There remains a gap in knowledge regarding the prevalence, penetrance, and manifestations of common hereditary cancer syndromes in the African-American population due to significant disparities in access and uptake of genetic testing. This review summarizes the available literature on genetic testing for breast, colon, and prostate cancers in the African-American population and explores the disparities in access to genetic testing between non-Hispanic White and African-American patients. This article also addresses the barriers to genetic testing and discrepancies in the uptake of recommendations for hereditary cancer syndromes in the African-American population when compared with non-Hispanic Whites. The review offers practice implications for many healthcare providers and demonstrates gaps in the existing knowledge to be addressed in future studies to help eliminate the persisting health disparities faced by the African-American population.
Collapse
Affiliation(s)
- Ambreen Khan
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Charles R Rogers
- Department of Family and Preventive Medicine, University of Utah School of Medicine, USA
| | - Carson D Kennedy
- Department of Family and Preventive Medicine, University of Utah School of Medicine, USA
| | - AnaMaria Lopez
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Joanne Jeter
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, Salt Lake City, UT, USA
| |
Collapse
|
50
|
Universal Immunohistochemistry for Lynch Syndrome: A Systematic Review and Meta-analysis of 58,580 Colorectal Carcinomas. Clin Gastroenterol Hepatol 2022; 20:e496-e507. [PMID: 33887476 DOI: 10.1016/j.cgh.2021.04.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Lynch syndrome is a form of hereditary colorectal cancer (CRC) caused by pathogenic germline variants (PV) in DNA mismatch repair (MMR) genes. Currently, many Western countries perform universal immunohistochemistry testing on CRC to increase the identification of Lynch syndrome patients and their relatives. For a clear understanding of health benefits and costs, data on its outcomes are required: proportions of Lynch syndrome, sporadic MMR-deficient (MMRd) cases, and unexplained MMRd cases. METHODS Ovid Medline, Embase, and Cochrane CENTRAL were searched for studies reporting on universal MMR immunohistochemistry, followed by MMR germline analysis, until March 20, 2020. Proportions were calculated, subgroup analyses were performed based on age and diagnostics used, and random effects meta-analyses were conducted. Quality was assessed using the Joanna Briggs Critical Appraisal Tool for Prevalence Studies. RESULTS Of 2723 identified articles, 56 studies covering 58,580 CRCs were included. In 6.22% (95% CI, 5.08%-7.61%; I2 = 96%) MMRd was identified. MMR germline PV was present in 2.00% (95% CI, 1.59%-2.50%; I2 = 92%), ranging from 1.80% to 7.27% based on completeness of diagnostics and age restriction. Immunohistochemistry outcomes were missing in 11.81%, and germline testing was performed in 76.30% of eligible patients. In 7 studies, including 6848 CRCs completing all diagnostic stages, germline PV and biallelic somatic MMR inactivation were found in 3.01% and 1.75%, respectively; 0.61% remained unexplained MMRd. CONCLUSIONS Age, completeness, and type of diagnostics affect the percentage of MMR PV and unexplained MMRd percentages. Complete diagnostics explain almost all MMRd CRCs, reducing the amount of subsequent multigene panel testing. This contributes to optimizing testing and surveillance in MMRd CRC patients and relatives.
Collapse
|