The effect of the Affordable Care Act on genetic testing patterns and outcomes for inheritable cancer syndromes: A single institution experience.

2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e13157-e13157
Author(s):  
Zhen Ni Zhou ◽  
Melissa Kristen Frey ◽  
Jessica Fields ◽  
Sushmita Gordhandas ◽  
Maria Paula Ruiz ◽  
...  

e13157 Background: National guidelines recommend genetic testing for women at increased risk for pathogenic mutations, however insurance coverage is a commonly cited barrier to test completion. The major provisions of the Affordable Care Act (ACA) became available in New York State (NYS) in January 2014. We sought to evaluate the effect of insurance status on genetic testing patterns following introduction of the ACA. Methods: Insurance status and genetic testing patterns at the hereditary breast and ovarian cancer center at a single institution were reviewed between 1/1/2013 and 12/31/2016. Insurance status and association with testing type and clinical outcomes were evaluated before and after January 2014. Results: During the study period, 1535 women underwent genetic counseling and genetic assessment. When comparing the cohort of women undergoing genetic testing prior to and following January 2014, significant increases in patients undergoing multigene panel testing were observed across all insurance types (6.3% vs. 40.6%, p < 0.001). While a significant increase in the number of patients with private insurance or Medicaid as primary coverage was seen, the increase in insured women was largely attributed to insurance polices through the ACA exchange (p < 0.001), with the majority being Medicaid ACA plans (80.3%). Conclusions: Since expansion of health insurance through the ACA in NYS, there has been a shift in the demographics of women undergoing genetic testing at a hereditary breast and ovarian cancer center with significantly more insured women and a rise in multigene panel testing. With insurance expansion and improved access to comprehensive genetic testing, insurance status should not be an obstacle for genetic testing.

2015 ◽  
Vol 81 (10) ◽  
pp. 941-944 ◽  
Author(s):  
Dt R. Howarth ◽  
Sharon S. Lum ◽  
Pamela Esquivel ◽  
Carlos A. Garberoglio ◽  
Maheswari Senthil ◽  
...  

Multigene panel testing for hereditary cancer risk has recently become commercially available; however, the impact of its use on patient care is undefined. We sought to evaluate results from implementation of panel testing in a multidisciplinary cancer center. We performed a retrospective review of consecutive patients undergoing genetic testing after initiating use of multigene panel testing at Loma Linda University Medical Center. From February 13 to August 25, 2014, 92 patients were referred for genetic testing based on National Comprehensive Cancer Network guidelines. Testing was completed in 90 patients. Overall, nine (10%) pathogenic mutations were identified: five BRCA1/2, and four in non-BRCA loci. Single-site testing identified one BRCA1 and one BRCA2 mutation. The remaining mutations were identified by use of panel testing for hereditary breast and ovarian cancer. There were 40 variants of uncertain significance identified in 34 patients. The use of panel testing more than doubled the identification rate of clinically significant pathogenic mutations that would have been missed with BRCA testing alone. The large number of variants of uncertain significance identified will require long-term follow-up for potential reclassification. Multigene panel testing provides additional information that may improve patient outcomes.


2019 ◽  
Vol 493 ◽  
pp. S576-S577
Author(s):  
V. Castillo-Guardiola ◽  
M.D. Sarabia-Meseguer ◽  
J.A. Macías-Cerrolaza ◽  
Á. García-Aliaga ◽  
L. Rosado-Jiménez ◽  
...  

Author(s):  
Eloise Chapman-Davis ◽  
Zhen Ni Zhou ◽  
Jessica C. Fields ◽  
Melissa K. Frey ◽  
Bailey Jordan ◽  
...  

2016 ◽  
Vol 34 (15_suppl) ◽  
pp. e13116-e13116
Author(s):  
Sarab Lizard ◽  
Marie Eliade ◽  
Jeremy Skrzypski ◽  
Amandine Baurand ◽  
Caroline Jacquot ◽  
...  

Oncotarget ◽  
2016 ◽  
Vol 8 (2) ◽  
pp. 1957-1971 ◽  
Author(s):  
Marie Eliade ◽  
Jeremy Skrzypski ◽  
Amandine Baurand ◽  
Caroline Jacquot ◽  
Geoffrey Bertolone ◽  
...  

2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 1533-1533
Author(s):  
Jessica Fields ◽  
Dimitrios Nasioudis ◽  
Zhen Ni Zhou ◽  
Ann Carlson ◽  
Melissa Kristen Frey ◽  
...  

1533 Background: Approximately one in forty Ashkenazi Jewish (AJ) individuals carry a BRCA1/2 mutation and genetic screening in this population has largely focused on these two genes. With the recent rapid uptake of multigene panel testing for cancer genetic assessment, we sought to explore multigene panels in our cohort which is comprised of AJ and non-AJ patients. Methods: The results of all patients with known ancestry who underwent genetic testing and counseling at the hereditary breast and ovarian cancer center at a single institution between 7/1/2013-12/31/2016 were reviewed. Results: One thousand six hundred and fifty patients with known ancestry underwent genetic testing over the study period, including 681 AJ patients. The median age was 49 (range 20-86). AJ patients were more likely to undergo targeted testing than non-AJ patients (74% vs. 61 %, P<0.001). The use of multigene panels in AJ patients increased over time (2013 – 3.2%, 2014 – 18.7%, 2015 – 27.4%, 2016 – 48.4%, P<0.001). Mutations were more common in AJ patients (75, 11% vs. 66, 7%, P=0.003). Variants of uncertain significance (VUS) were less common in AJ patients (40, 6% vs. 124, 13%, P<0.001), even when excluding patients with single gene testing (32, 19% vs. 98, 27%, P=0.05). Among all patients, mutations in BRCA1/2 were most common (75%). The majority (69%) of non- BRCA1/2 mutations were identified on multigene panels. Rates of mutations in non- BRCA1/2 genes were the same among AJ and non-AJ patients (16, 21% vs. 20, 30%, P=0.3, Table 1). Conclusions: AJ patients have equivalent rates of non- BRCA1/2 mutations and on multigene panels have lower rates of VUS compared to non-AJ patients. However, the majority of AJ patients underwent targeted gene testing. These findings suggest consideration of a change in paradigm for genetic assessment of AJ patients with a focus on BRCA and non- BRCAassociated cancer genes through multigene panel testing. [Table: see text]


2021 ◽  
Vol 10 (3) ◽  
pp. 207-217
Author(s):  
Stacey Dacosta Byfield ◽  
Helen Wei ◽  
Mary DuCharme ◽  
Johnathan M Lancaster

Aim: Healthcare utilization and costs were compared following 25-gene panel (panel) or single syndrome (SS) testing for hereditary breast and ovarian cancer. Materials & methods: Retrospective cohort study of patients unaffected by cancer with panel (n = 6359) or SS (n = 4681) testing for hereditary breast and ovarian cancer (01 January 2014 to 31 December 2016). Groups were determined by test type and result (positive, negative, variant of uncertain significance [VUS]). Results: There were no differences in total unadjusted healthcare costs between the panel (US$14,425) and SS (US$14,384) groups (p = 0.942). Among VUS patients in the panel and SS groups, mean all-cause costs were US$14,404 versus US$20,607 (p = 0.361) and mean risk-reduction/early detection-specific costs were US$718 versus US$679 (p = 0.890), respectively. Adjusted medical costs were not significantly different between panel and SS cohorts. Conclusion: Healthcare utilization and costs were comparable between the SS and panel tests overall and for patients with VUS.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 1585-1585
Author(s):  
Chethan Ramamurthy ◽  
Mark A. Hitrik ◽  
Lyudmila DeMora ◽  
Andrea Forman ◽  
Kim Rainey ◽  
...  

1585 Background: Genetic testing for hereditary cancer predisposition has rapidly changed over the past few years with the introduction of multigene panel testing. Multigene testing has evolved from disease-agnostic comprehensive (C) panels alone to include disease-specific but expanded (DSE) panels as well as guideline-based (GB) panels. We analyzed trends in utilization of genetic testing over a two-year period in one NCI-designated Cancer Center, hypothesizing that over time genetic testing usage would trend toward more disease-specific panels. Methods: We conducted a retrospective analysis of our program’s database for all germline genetic tests ordered from 9/1/2013 to 8/31/2015 (n = 619; 246 in year 1, and 373 in year 2). Tests were categorized into three groups based on specificity: GB (range: 2-12 genes tested), DSE (12-35 genes tested), and C (28-80 genes tested). The Chi-square test was used to analyze test types ordered in year 1 (9/1/2013-8/31/2014) and year 2 (9/1/2014 – 8/31/2015) and the proportions of resulting mutation types. Results: A total of 604 germline genetic tests met the inclusion criteria: 39 GB (20 year 1, 19 year 2), 171 DSE (43 year 1, 128 year 2), and 394 C (180 year 1, 214 year 2). Compared to year 1, a larger proportion of DSE tests (35% v. 18%, p < 0.001), and a smaller proportion of C tests (59% v. 74%, p < 0.001) and GB tests (5% vs. 8%, p = 0.146) were ordered. DSE panels revealed a pathogenic variant (PV) at a rate of 16% and a variant of unknown significance (VUS) at a rate of 24%. C tests revealed a PV and VUS at rates of 14% and 29%, respectively. GB tests revealed a PV and VUS at rates of 21% and 18%, respectively. No statistically significant differences in detection rates of mutation types (PV or VUS) were found between GB, DSE, or C tests. Conclusions: The rates of PV detection were not significantly different between test types, but the profile of tests ordered changed over time to favor DSE panels. Exploration of factors contributing to changing trends in genetic testing are warranted as counselors and clinicians adapt to the quickly expanding number of genes associated with hereditary cancer risks, many of them moderate-risk, and the evolving landscape of multigene panel testing.


Sign in / Sign up

Export Citation Format

Share Document