Background
Ovarian cancer (OC) is the most common cause of gynecologic cancer deaths worldwide annually (324,603 new cases, 206,956 deaths),1 with approximately 90% of cases being epithelial OC.2 By 2045, OC cases are predicted to increase by 24% in the United Kingdom, 28% in the United States, and 47% worldwide.1 Germline pathogenic and likely pathogenic variants (henceforth termed “pathogenic variants” [PVs]) in BRCA1/BRCA2 comprise most of the known inheritable component of OC risk and are found in 10% to 15% of epithelial OC.3–5 BRCA1/BRCA2 PVs are associated with a 17% to 44% OC risk and a 69% to 72% breast cancer (BC) risk by 80 years of age.6 PARP inhibitor (PARPi) therapy is recommended for patients with OC with germline or somatic BRCA1/2 PVs because it increases overall survival (OS) and progression-free survival (PFS) in both primary and recurrence settings.7–13 Determining BRCA status helps decide treatment options, with olaparib being the first PARPi recommended for first-line maintenance treatment of platinum-sensitive, BRCA-mutated advanced OC.14 However, approximately 50% of BRCA PVs are missed by traditional family history (FH)–based testing.4,15–17 Guidelines now recommend mainstreaming unselected BRCA testing at OC diagnosis for initially germline18 and subsequently somatic testing.19–21 Lately, women with other cancer-susceptibility genes (CSGs) in the homologous recombination repair pathway, such as RAD51C, RAD51D, and BRIP1, with validated moderate lifetime OC risks of 5.8% to 13%22,23 are being offered surgical prevention.24–27 Testing for these CSGs of clinical utility28 can enable wider therapeutic benefit and is now recommended. Although CSG testing at OC diagnosis has been driven by increasing applicability for therapeutic oncology, arguably the major impact on disease burden overall may come from opportunities for precision prevention. Unselected multigene panel germline testing itself can, through cascade testing, identify more unaffected relatives with PVs who can benefit from BC/OC screening and prevention, as well as identify women with OC themselves who can benefit from screening and prevention of secondary BC. Wide implementation and sustainability of changes in clinical practice requires that they be cost-effective for the health system. Unselected BRCA testing at OC diagnosis is cost-effective compared with no testing, but comparison with the clinical comparator of clinical criteria/FH-based testing is lacking, and these earlier analyses excluded PARPi treatment.29 Both PARPi costs and OS results are critical parameters affecting cost-effectiveness. However, cost-effectiveness data on multigene panel germline testing at OC diagnosis are lacking. Additionally, the cost-effectiveness of parallel panel germline and somatic testing has not yet been established.
Using data from 4 OC cohorts in the UK and US along with modeling, we estimated, for the first time, the incremental lifetime effects, costs, and cost-effectiveness of parallel panel germline and somatic BRCA testing of all patients with OC compared with the earlier standard of clinical criteria/FH-based genetic (BRCA) testing in the UK and US health systems. Our analysis incorporates PARPi therapy and explores a range of PARPi costs and OS results to establish thresholds for the cost-effectiveness of this important clinical strategy. We also compared unselected panel BRCA1/BRCA2/RAD51C/RAD51D/BRIP1 germline testing itself with clinical criteria/FH-based BRCA testing to evaluate the potential benefit from unselected panel germline testing.
Methods
We obtained CSG and FH data by age from 2,391 “unselected” patients with OC from 4 cohort studies: (1) North-East London Cancer Network cohort (n=298 from the SIGNPOST study [ISRCTN-16988857])4; (2) Manchester cohort (Manchester University NHS Foundation Trust; n=751)30; (3) Scottish cohort (n=434)31; and (4) Washington cohort (n=908 from University of Washington Medical Center). We obtained the proportion fulfilling standard FH/clinical criteria for genetic testing (henceforth termed FH-positive) by age group (see Table S1 in the supplementary material, available online with this article) and CSG PV prevalence among unselected OC cases in each setting. We obtained population-based OC incidence by age from Cancer Research UK 201532 (UK analysis) and US Cancer Statistics 201533 (US analysis). From this, we calculated the total FH-positive and CSG PV-positive OC cases depending on the annually newly diagnosed OC cases by age group in UK/US women (Supplementary Table S1).
Model and Testing Strategy
Using TreeAge Pro software, we developed an individual-level microsimulation model (Supplementary Figure S1) to analyze the lifetime costs and health effects of parallel BRCA1/BRCA2/RAD51C/RAD51D/BRIP1 panel germline testing and BRCA1/BRCA2 somatic testing of all patients with OC incorporating PARPi therapy (strategy A). This strategy was compared with the historical clinical comparator of clinical criteria/FH-based BRCA1/BRCA2 germline testing (strategy B). Because unselected multigene panel germline testing itself can identify more unaffected relatives with PVs who can undergo risk-reducing interventions, we also compared unselected panel BRCA1/BRCA2/RAD51C/RAD51D/BRIP1 germline testing alone (excluding somatic testing and PARPi treatment) with strategy B. Additionally, we compared strategy A with unselected BRCA germline testing in a scenario analysis. In strategy A, all patients underwent counseling and panel germline and somatic testing. In strategy B, only those fulfilling clinical/FH criteria underwent counseling and BRCA germline testing. For the base case, we presumed that all eligible patients underwent genetic testing. If patients had a BRCA1/BRCA2/RAD51C/RAD51D/BRIP1 PV, their first-degree relatives (FDR) were tested for the familial PV, and the second-degree relatives were tested if the FDR was found to have a BRCA1/BRCA2/RAD51C/RAD51D/BRIP1 PV. We assumed that all eligible relatives were tested in the base case but also undertook a scenario analysis with lower (70%) uptake of cascade testing. We incorporated an 8.8% variant of uncertain significance (VUS) rate (BRCA1/BRCA2, 4.86%; RAD51C/RAD51D/BRIP1, 3.93%)4,30,31 and an 8.7% pathogenic/likely pathogenic VUS reclassification rate.34
Unaffected BRCA1/BRCA2/RAD51C/RAD51D/BRIP1 PV carriers could choose risk-reducing salpingo-oophorectomy (RRSO) to reduce their OC risk,35,36 and unaffected BRCA1/BRCA2 PV carriers could choose risk-reducing mastectomy (RRM)37 or chemoprevention with selective estrogen receptor modulators (SERM) for BC risk reduction38 and MRI-based/mammography-based enhanced BC screening. OC cases with germline/somatic BRCA1/BRCA2 PVs could opt for PARPi therapy. We assumed that 71% of patients with BRCA-mutated OC received PARPi therapy given that 88% of patients with BRCA-mutated OC have been shown to experience a response to first-line platinum-based chemotherapy,39 and 81% have been shown to present in advanced stages.40
Although initial studies suggested that premenopausal RRSO reduced BC risk,36,41,42 more recent data contradict this.43–45 Hence, conservatively, we assumed no BC risk reduction from RRSO. We included an excess risk and mortality from coronary heart disease (CHD) in premenopausal women who do not take hormone-replacement therapy (HRT) following RRSO (absolute mortality increase, 3.03%).46,47 Patients with OC and their cancer-free relatives may pass through various health states in the model: no cancer, sporadic OC, germline OC, somatic OC, sporadic BC, germline BC, and both BC and OC. Cancer incidence was determined by summing the probabilities of pathways ending in OC or BC. The potential population impact was estimated from the additional reduction in BC and OC incidence following testing of all OC cases occurring annually in UK/US women.
Probabilities
Model pathway probabilities are provided in Supplementary Table S2. The age-specific general population BC/OC incidences were obtained from Cancer Research UK 201532,48 (UK analysis) and US Cancer Statistics 201533 (US analysis), and BC/OC incidence for BRCA1/BRCA2 carriers were obtained from the literature.6 RAD51C confers an increased relative risk of 7.55 (CI, 5.60-10.19),23 RAD51D a relative risk of 7.60 (CI, 5.61–0.30),23 and BRIP1 a relative risk of 3.41 (CI, 2.12–5.54).22
Number and Age Distribution of Relatives
The new OC cases by age groups in the UK and US calibrated the age distribution of patients in the model. The Office for National Statistics (UK)49 and the National Center for Health Statistics (US)50 data helped estimate FDRs/second-degree relatives and their ages relative to index cases for UK and US women, respectively (see Supplementary Table S3). Lifetables helped estimate probabilities for relatives at different ages being alive and to compute the age distribution and number of relatives that undergo genetic testing.
Costs
Costs are reported as 2019 prices. Both payer/societal-perspective analyses were undertaken. We included costs of germline testing, somatic testing, pretest and posttest genetic counseling,51,52 BC and OC treatment, excess CHD, and productivity loss. UK costs were obtained from National Health Service (NHS) reference costs53,54 and converted wherever needed using the Hospital and Community Health Services Index.55 US costs from the literature were inflated to 2019 US dollars using the medical component of the Consumer Price Index. The list-price cost of olaparib (PARPi) was £2,317.5 per 14-day pack (UK) and $13,886 per 30-day pack (US).14,56 Germline testing cost was £150/$200 and somatic testing cost was £360/$480. As per National Institute for Health and Care Excellence (NICE) recommendations, future health care costs not associated with BC/OC/CHD were excluded.57 See Supplementary Table S4 for an explanation of costs, and Supplementary Methods S1 for costs from productivity loss.
Life Years
Our analysis incorporates a lifetime time horizon, and relevant lifetables estimate life expectancy in unaffected women. See Supplementary Methods S2 for survival estimates. We assumed that the median age was 37 years for RRM and 40 years for RRSO.58 BC and OC survival were modeled using 5-year survival data from the global surveillance of cancer survival.59 No significant overall long-term survival differences between germline and sporadic BC/OC have been found.40,60,61 Patients with OC with germline/somatic BRCA receiving first-line PARPi therapy have improved OS (hazard ratio [HR], 0.55; CI, 0.40–0.76),13 and we additionally explored its uncertainty through a range of scenario and sensitivity analyses.
Quality-Adjusted Life Years
NICE recommends quality-adjusted life years (QALYs) for measuring health outcomes. See Supplementary Methods S3 for QALYs/utility scores within the model.
Statistical Analysis
ICERs obtained were compared with presumed willingness-to-pay (WTP) thresholds: UK analysis = £30,000/QALY63 and US analysis = $100,000/QALY.64 We evaluated the cost-effectiveness of unselected panel germline testing alone (without somatic testing/PARPi) compared with FH-based BRCA testing through a scenario analysis. We undertook a number of other scenario analyses: (1) half HRT compliance (40%) with and without PARPi therapy; (2) lower uptake rate (70%) of germline testing in unaffected relatives; (3) parallel germline and somatic testing in patients aged <70 years, and sequential somatic testing followed by germline testing if somatic PV was identified in patients aged ≥70 years as recent data highlight this possibility65; (4) 50% reduced RRM/RRSO rates; and (5) comparison of panel germline and BRCA somatic testing (strategy A) with unselected BRCA germline testing. Additionally, we evaluated the maximum cost(s) of PARPi therapy to maintain cost-effectiveness of offering panel germline and BRCA somatic testing (strategy A) across various OC survival estimates, where ICERs of strategy A equal the WTP thresholds in the UK and US, respectively.
Wide-ranging one-way and probabilistic sensitivity analyses (PSAs) were undertaken to evaluate model uncertainty. Model parameters are varied individually in one-way analyses and simultaneously in the PSAs.63 Probabilities/utility scores were varied by their 95% confidence intervals/range or by ±10% and costs by ±30%. Costs were given a γ distribution, quality-of-life was given a log-normal distribution, and probability was given a β distribution, as recommended.66 For PSAs, we obtained 1,000 estimates of incremental costs and effects by sampling from the distributions of each parameter. Cost-effectiveness acceptability curves demonstrated whether (1) panel germline and BRCA somatic testing with PARPi treatment and (2) panel germline testing alone (without somatic testing/PARPi treatment) for all patients with OC are cost-effective across varying WTP thresholds.
Results
Overall lifetime costs, QALYs, and ICERs for UK/US women are presented in Table 1. Unselected parallel panel germline and BRCA somatic testing with PARPi therapy for all patients with OC diagnosed annually (strategy A) compared with clinical criteria/FH-based BRCA testing was not cost-effective in the base-case analysis. UK ICERs were £51,175/QALY from the payer perspective and £50,202/QALY from the societal perspective, and US ICERs were $175,232/QALY from the payer perspective and $174,667/QALY from the societal perspective. However, unselected panel germline testing is cost-effective compared with FH-based BRCA testing (without PARPi therapy). UK ICERs were £11,291/QALY from the payer perspective and £6,923/QALY from the societal perspective, and US ICERs were $68,808/QALY from the payer perspective and $65,786/QALY from the societal perspective. This would remain cost-effective even if genetic testing costs increased to £1,321/£1,594 (UK payer perspective/societal perspective) or $1,626/$1,765 (US payer perspective/societal perspective). Parallel panel germline and BRCA somatic testing with PARPi therapy was not cost-effective compared with unselected BRCA germline testing alone, with UK ICERs being £105,934/QALY from the payer perspective and £105,433/QALY from the societal perspective, and US ICERs being $553,422/QALY from the payer perspective and $553,240/QALY from the societal perspective. Strategy A is extremely sensitive to both PARPi costs and OS estimates from PARPi treatment. Panel germline and somatic testing with PARPi (strategy A) can become cost-effective for both the UK and the US if the OS HR improves from 0.55 (base case) to 0.28. The yearly PARPi list price is £60,462 in the UK, and $169,067 in the US. Strategy A becomes cost-effective if annual PARPi treatment costs decrease by 45% (UK cost, £33,006) or 46% (US cost, $90,841). The maximum PARPi costs for strategy A to remain cost-effective at different OS HRs (0.3–0.7) from payer/societal perspectives for the UK and US (see Figure 1) show that the HR for OS is inversely related to PARPi costs. Annual PARPi costs from payer/societal perspectives need to decrease to £24,030/£25,565 in the UK and $54,438/$55,042 in the US if the OS HR is 0.7. Various scenario analyses are illustrated in Table 1.
Lifetime Discounted Costs and Effects per Woman, ICER of Panel Germline Testing, and Somatic BRCA Testing for All Patients With OC
Maximum yearly PARP costs to remain cost-effective. The yearly cost of PARPi therapy is £60,462 in the UK and $169,067 in the US in the base-case analysis. The maximum yearly PARPi costs for unselected panel germline and somatic testing with PARPi therapy to remain cost-effective from the payer and societal perspectives, at WTP thresholds of (A) £30,000/QALY in the UK and (B) $100,000/QALY in the US. Different scenarios for the HR for ovarian cancer survival from PARPi were explored, ranging from 0.3 to 0.7.
Abbreviations: HR, hazard ratio; PARPi, PARP inhibitor; QALY, quality-adjusted life year; WTP, willingness-to-pay.
Citation: Journal of the National Comprehensive Cancer Network 22, 2D; 10.6004/jnccn.2023.7331
The population effects of reductions in BC/OC incidence and deaths are presented in Table 2. The unaffected female relative PV carriers identified through cascade testing were 1.41 (UK) and 1.49 (US) per index PV carrier with OC (see Supplementary Table S3). Unselected panel germline and somatic testing (strategy A) can lead to an average additional 348-day increase in life expectancy for UK CSG PV carriers (397-day increase for PV carrier patients and 322-day increase for PV carrier unaffected relatives) and 278 days for US CSG PV carriers (380-day increase for PV carrier patients and 207-day increase for PV carrier unaffected relatives). For unaffected relatives identified as PV carriers, those who underwent RRM or RRSO had 529-day (UK) and 445-day (US) increases in life expectancy compared with those who did not undergo RRM or RRSO. One year’s unselected panel germline and somatic testing for all patients with OC could prevent an additional 171 BC cases and 38 OC cases in UK women and 461 BC cases and 99 OC cases in US women (Table 2). Annually, strategy A translates to averting 192 UK cancer deaths and 460 US cancer deaths across a lifetime horizon (Table 2).
Lifetime Population Impact of Multigenetic Testing and Somatic BRCA Testing for All Patients With OC
The PSA results (Figure 2) show that unselected panel germline testing and BRCA1/BRCA2 somatic testing for patients with OC incorporating PARPi is cost-effective at the WTP thresholds for 29% (UK payer), 4% (US payer), or 8% (US societal) of simulations. However, unselected panel germline testing alone without PARPi therapy is cost-effective at the WTP thresholds for 99% (UK payer), 96% (US payer), and 100% (US societal) of simulations.
Probabilistic sensitivity analysis results. All model parameters/variables are varied simultaneously across their distributions to further explore model uncertainty in probabilistic sensitivity analysis. The results of 1,000 simulations were plotted on a cost-effectiveness acceptability curve showing the proportion of simulations (y axis) that indicated that the intervention was cost-effective at different WTP thresholds (x axis). Results are presented for comparison of both strategies: parallel panel germline and somatic testing with PARPi from the (A) UK payer perspective, (B) US payer perspective, and (C) US societal perspective, and panel germline testing without somatic testing or PARPi from the (D) UK payer perspective, (E) US payer perspective, and (F) US societal perspective.
Abbreviations: PARPi, PARP inhibitor; QALY, quality-adjusted life year; WTP, willingness-to-pay.
Citation: Journal of the National Comprehensive Cancer Network 22, 2D; 10.6004/jnccn.2023.7331
One-way sensitivity analyses (Supplementary Figure S2) show that PARPi cost is the main variable having the biggest impact on the cost-effectiveness results, and OS is also important. Without PARPi therapy, individual variables, such as PV prevalence, costs, utility scores, and transition probabilities, have very minimal impact on the cost-effectiveness of unselected panel germline testing.
Discussion
We show, for the first time, that offering unselected parallel panel germline testing and somatic BRCA1/BRCA2 testing for patients with OC incorporating PARPi therapy has higher ICERs than the established cost-effectiveness thresholds for UK/US health systems. However, this can become cost-effective if PARPi treatment costs decrease by 45% in the UK and 46% in the US, or if the final OS following PARPi treatment reaches an HR of 0.28 rather than the established base-case HR of 0.55. This is critically important because implementation of such a program has significant clinical benefit, leading annually to 209 fewer cases of and 192 fewer deaths from BC and OC in UK women and 560 fewer cases of and 460 fewer deaths from BC and OC in US women.
Notably, unselected panel germline testing for patients with OC alone (excluding PARPi) is cost-effective, with ICERs well below considered WTP thresholds. This remains cost-effective even at higher genetic costs of up to £1,321 to £1,594 or $1,626 to $1,765 (well within the costs of most providers) and even if RRM or RRSO rates decline by 50%. Our results support unselected panel germline testing at OC diagnosis, which can identify 3% to 4% more PV carriers (compared with BRCA testing alone) who can benefit from precision prevention.4,5 Most current guidelines advocate BRCA testing at OC diagnosis only.19,21 It is important that these recommendations are expanded to include a panel of OC genes that have clear clinical utility. Besides RAD51C/RAD51D/BRIP1 genes, a recommended OC panel should also include moderate-risk PALB267 and Lynch syndrome genes found in 1% patients with OC.68–70 This can provide greater stimulus for early diagnosis/prevention in unaffected family members, preventing more cancers and saving more lives.
Earlier studies demonstrating the cost-effectiveness of germline BRCA1/BRCA2 testing in patients with OC29,71 compared unselected genetic testing with no testing, rather than clinical criteria/FH-based testing, which is a better clinical comparator. Our study uses a more appropriate comparator for evaluating cost-effectiveness. Our study also used a large sample of population-based UK and US patients with OC and is broader in scope through incorporating more ovarian CSGs (RAD51C/RAD51D/BRIP1), somatic BRCA1/BRCA2 testing, and PARPi treatment. Prior PARPi cost-effectiveness studies have predominantly evaluated its use in a recurrent (not first-line) setting and used surrogate outcomes, such as progression-free life years or progression-free QALYs, to draw conclusions.72 However, there are no theoretical or empirical thresholds for cost-effectiveness using PFS as an effectiveness measure, and thus it is incorrect to draw conclusions on PARPi cost-effectiveness in this manner. Most studies suggest that PARPi is not cost-effective as maintenance therapy for platinum-sensitive recurrent OC, with high drug acquisition costs73,74 being a major factor. An initial health technology assessment evaluation by NICE following a pharmaceutical company submission indicated that olaparib was not cost-effective for first-line maintenance treatment of BRCA-mutated OC, though it could potentially become cost-effective in the future.14 The NICE evidence review group highlighted the significant uncertainty and potential overestimation of OS, the overestimation of eligibility, and limited flexibility of costs, leading to ICERs higher than the current WTP threshold. The group concluded that NICE was unable to recommend olaparib for routine NHS use but supported its use through the Cancer Drugs Fund pending OS results given its future cost-effectiveness potential.14 Our results lend further credence to high PARPi costs being a major factor in determining cost-effectiveness, as evidenced from the one-way sensitivity analysis and hugely different ICERs in the scenarios with and without PARPi therapy (Table 1).
We evaluated unselected parallel germline panel testing and somatic BRCA testing because this approach arguably maximizes PV identification for patient benefit and precision prevention.4 We preferred this to a sequential somatic first strategy because somatic testing may miss large genomic rearrangements, which in some populations (including in the UK) can comprise approximately 10% of PVs.4 This parallel approach is recommended in UK guidelines and is part of routine NHS care.75 However, there may be countries or populations where LGR rates are negligible or very low. These jurisdictions may choose to have a sequential somatic first (followed by germline) approach to mainstreaming genetic testing.
Our study has many strengths, including drawing data form large population-based cohorts and adhering to NICE recommendations of cost utility analysis for economic evaluation.57 We used QALYs for health outcomes, discount for costs/outcomes, presented a lifetime horizon, performed extensive sensitivity and scenario analyses to support the strength/accuracy of results, covered societal/payer perspectives, incorporated a detriment for CHD mortality,46 and detailed a comprehensive range of costs. We used the most recently published OS estimates from olaparib therapy (HR, 0.55; CI, 0.40–0.76)13 instead of the earlier surrogate of OS (progression-free survival 2 [PFS-2], defined as the time from randomization to progression on first subsequent therapy) due to the immature clinical effectiveness data. We also provide the maximum PARPi costs to maintain cost-effectiveness at different HRs of OS, which is useful for providers and decision-makers.
A potential limitation of our analysis was the exclusion of HRD testing; however, HRD tests are extremely expensive, making this approach not cost-effective.76 They are not universally available or implemented, and the SOLO-1 study,13 whose survival data were used in our analysis, did not include HRD testing. We have also not evaluated the combination of PARPi with other drugs or agents,77 which will need to be explored in other studies.
Randomized trial results have led to the US FDA, European Medicines Agency, and other countries approving PARPi for first-line maintenance treatment of BRCA-mutated advanced OC, bringing about a paradigm shift in the clinical management of this population of women. The skyrocketing costs of new oncology drugs, leading to financial toxicity, restricted availability, rising out-of-pocket costs, and inequality in access among patients, has become a major global problem.78,79 For widescale implementation and equitable access, it is important that new drugs are priced at a level that is cost-effective and affordable for health systems. Our analysis highlighting potential cost-effective price thresholds for olaparib is an important pointer in this regard. For broadening equity and access, even lower price thresholds will be needed for middle- and lower-income countries.
Conclusions
Our findings suggest that unselected panel germline and somatic testing for patients with OC can substantially reduce future BC and OC cases and related deaths compared with a clinical criteria/FH-based strategy. This approach can become cost-effective if PARPi costs decrease by 45% to 46%. Nevertheless, panel germline testing alone is highly cost-effective and maximizes variant identification for precision prevention. It is important for clinical germline/genetic testing guidelines to move from single-gene (BRCA1/2) testing toward a multigene panel testing approach.
References
- 1.↑
International Agency for Research on Cancer. Cancer tomorrow. Accessed January 20, 2019. Available at: https://gco.iarc.who.int/media/globocan/factsheets/cancers/25-ovary-fact-sheet.pdf
- 2.↑
Cheung A, Shah S, Parker J, et al. Non-epithelial ovarian cancers: how much do we really know? Int J Environ Res Public Health 2022;19:1106.
- 3.↑
Zhang S, Royer R, Li S, et al. Frequencies of BRCA1 and BRCA2 mutations among 1,342 unselected patients with invasive ovarian cancer. Gynecol Oncol 2011;121:353–357.
- 4.↑
Chandrasekaran D, Sobocan M, Blyuss O, et al. Implementation of multigene germline and parallel somatic genetic testing in epithelial ovarian cancer: SIGNPOST study. Cancers (Basel) 2021;13:4344.
- 5.↑
Norquist BM, Harrell MI, Brady MF, et al. Inherited mutations in women with ovarian carcinoma. JAMA Oncol 2016;2:482–490.
- 6.↑
Kuchenbaecker KB, Hopper JL, Barnes DR, et al. Risks of breast, ovarian, and contralateral breast cancer for BRCA1 and BRCA2 mutation carriers. JAMA 2017;317:2402–2416.
- 7.↑
Ledermann J, Harter P, Gourley C, et al. Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial. Lancet Oncol 2014;15:852–861.
- 8.↑
Moore K, Colombo N, Scambia G, et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med 2018;379:2495–2505.
- 9.↑
Coleman RL, Oza AM, Lorusso D, et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;390:1949–1961.
- 10.↑
Pujade-Lauraine E, Ledermann JA, Selle F, et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol 2017;18:1274–1284.
- 11.↑
Schettini F, Giudici F, Bernocchi O, et al. Poly (ADP-ribose) polymerase inhibitors in solid tumours: systematic review and meta-analysis. Eur J Cancer 2021;149:134–152.
- 12.↑
Banerjee S, Moore KN, Colombo N, et al. Maintenance olaparib for patients with newly diagnosed advanced ovarian cancer and a BRCA mutation (SOLO1/GOG 3004): 5-year follow-up of a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2021;22:1721–1731.
- 13.↑
DiSilvestro P, Banerjee S, Colombo N, et al. Overall survival with maintenance olaparib at a 7-year follow-up in patients with newly diagnosed advanced ovarian cancer and a BRCA mutation: the SOLO1/GOG 3004 trial. J Clin Oncol 2023;41:609–617.
- 14.↑
National Institute for Health and Care Excellence. Olaparib for maintenance treatment of BRCA mutation-positive advanced ovarian, fallopian tube or peritoneal cancer after response to first-line platinum-based chemotherapy. Accessed September 25, 2019. Available at: https://www.nice.org.uk/guidance/ta598/evidence
- 15.↑
George A, Riddell D, Seal S, et al. Implementing rapid, robust, cost-effective, patient-centred, routine genetic testing in ovarian cancer patients. Sci Rep 2016;6:29506.
- 16.↑
Møller P, Hagen AI, Apold J, et al. Genetic epidemiology of BRCA mutations—family history detects less than 50% of the mutation carriers. Eur J Cancer 2007;43:1713–1717.
- 17.↑
Alsop K, Fereday S, Meldrum C, et al. BRCA mutation frequency and patterns of treatment response in BRCA mutation-positive women with ovarian cancer: a report from the Australian Ovarian Cancer Study Group. J Clin Oncol 2012;30:2654–2663.
- 18.↑
NHS England. Clinical commissioning policy: genetic testing for BRCA1 and BRCA2 mutations. Accessed May 1, 2015. Available at: https://www.engage.england.nhs.uk/consultation/specialised-services-consultation/user_uploads/brca-policy.pdf
- 19.↑
Konstantinopoulos PA, Norquist B, Lacchetti C, et al. Germline and somatic tumor testing in epithelial ovarian cancer: ASCO guideline. J Clin Oncol 2020;38:1222–1245.
- 20.↑
Miller RE, Leary A, Scott CL, et al. ESMO recommendations on predictive biomarker testing for homologous recombination deficiency and PARP inhibitor benefit in ovarian cancer. Ann Oncol 2020;31:1606–1622.
- 21.↑
Sundar S, Manchanda R, Gourley C, et al. British Gynaecological Cancer Society/British Association of Gynaecological Pathology consensus for germline and tumor testing for BRCA1/2 variants in ovarian cancer in the United Kingdom. Int J Gynecol Cancer 2021;31:272–278.
- 22.↑
Ramus SJ, Song H, Dicks E, et al. Germline mutations in the BRIP1, BARD1, PALB2, and NBN genes in women with ovarian cancer. J Natl Cancer Inst 2015;107:djv214.
- 23.↑
Yang X, Song H, Leslie G, et al. Ovarian and breast cancer risks associated with pathogenic variants in RAD51C and RAD51D. J Natl Cancer Inst 2020;112:1242–1250.
- 24.↑
Manchanda R, Legood R, Antoniou AC, et al. Specifying the ovarian cancer risk threshold of ‘premenopausal risk-reducing salpingo- oophorectomy’ for ovarian cancer prevention: a cost-effectiveness analysis. J Med Genet 2016;53:591–599.
- 25.↑
Manchanda R, Menon U. Setting the threshold for surgical prevention in women at increased risk of ovarian cancer. Int J Gynecol Cancer 2018;28:34–42.
- 26.↑
Manchanda R, Gaba F, Talaulikar V, et al. Risk-reducing salpingo-oophorectomy and the use of hormone replacement therapy below the age of natural menopause: scientific impact paper no. 66 October 2021: scientific impact paper no. 66. BJOG 2022;129:e16–34.
- 27.↑
Hanson H, Kulkarni A, Loong L, et al. UK consensus recommendations for clinical management of cancer risk for women with germline pathogenic variants in cancer predisposition genes: RAD51C, RAD51D, BRIP1 and PALB2. J Med Genet 2023;60:417–429.
- 28.↑
Domchek SM, Robson ME. Update on genetic testing in gynecologic cancer. J Clin Oncol 2019;37:2501–2509.
- 29.↑
Eccleston A, Bentley A, Dyer M, et al. A cost-effectiveness evaluation of germline BRCA1 and BRCA2 testing in UK women with ovarian cancer. Value Health 2017;20:567–576.
- 30.↑
Morgan RD, Burghel GJ, Flaum N, et al. Prevalence of germline pathogenic BRCA1/2 variants in sequential epithelial ovarian cancer cases. J Med Genet 2019;56:301–307.
- 31.↑
Rust K, Spiliopoulou P, Tang CY, et al. Routine germline BRCA1 and BRCA2 testing in patients with ovarian carcinoma: analysis of the Scottish real-life experience. BJOG 2018;125:1451–1458.
- 32.↑
Cancer Research UK. Ovarian cancer incidence statistics. Accessed July 5, 2020. Available at: http://www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/ovarian-cancer/incidence#heading-One
- 33.↑
Centers for Disease Control and Prevention. Rate of new cancers by age group: all races, female. Accessed November 19, 2018. Available at: https://gis.cdc.gov/Cancer/USCS/DataViz.html
- 34.↑
Mersch J, Brown N, Pirzadeh-Miller S, et al. Prevalence of variant reclassification following hereditary cancer genetic testing. JAMA 2018;320:1266–1274.
- 35.↑
Finch A, Beiner M, Lubinski J, et al. Salpingo-oophorectomy and the risk of ovarian, fallopian tube, and peritoneal cancers in women with a BRCA1 or BRCA2 mutation. JAMA 2006;296:185–192.
- 36.↑
Rebbeck TR, Kauff ND, Domchek SM. Meta-analysis of risk reduction estimates associated with risk-reducing salpingo-oophorectomy in BRCA1 or BRCA2 mutation carriers. J Natl Cancer Inst 2009;101:80–87.
- 37.↑
Rebbeck TR, Friebel T, Lynch HT, et al. Bilateral prophylactic mastectomy reduces breast cancer risk in BRCA1 and BRCA2 mutation carriers: the PROSE study group. J Clin Oncol 2004;22:1055–1062.
- 38.↑
Cuzick J, Sestak I, Bonanni B, et al. Selective oestrogen receptor modulators in prevention of breast cancer: an updated meta-analysis of individual participant data. Lancet 2013;381:1827–1834.
- 39.↑
Vencken PM, Kriege M, Hoogwerf D, et al. Chemosensitivity and outcome of BRCA1- and BRCA2-associated ovarian cancer patients after first-line chemotherapy compared with sporadic ovarian cancer patients. Ann Oncol 2011;22:1346–1352.
- 40.↑
McLaughlin JR, Rosen B, Moody J, et al. Long-term ovarian cancer survival associated with mutation in BRCA1 or BRCA2. J Natl Cancer Inst 2013;105:141–148.
- 41.↑
Chai X, Domchek S, Kauff N, et al. RE: breast cancer risk after salpingo-oophorectomy in healthy BRCA1/2 mutation carriers: revisiting the evidence for risk reduction. J Natl Cancer Inst 2015;107:djv217.
- 42.↑
Domchek SM, Friebel TM, Singer CF, et al. Association of risk-reducing surgery in BRCA1 or BRCA2 mutation carriers with cancer risk and mortality. JAMA 2010;304:967–975.
- 43.↑
Mavaddat N, Antoniou AC, Mooij TM, et al. Risk-reducing salpingo-oophorectomy, natural menopause, and breast cancer risk: an international prospective cohort of BRCA1 and BRCA2 mutation carriers. Breast Cancer Res 2020;22:8.
- 44.↑
Heemskerk-Gerritsen BA, Seynaeve C, van Asperen CJ, et al. Breast cancer risk after salpingo-oophorectomy in healthy BRCA1/2 mutation carriers: revisiting the evidence for risk reduction. J Natl Cancer Inst 2015;107:djv033.
- 45.↑
Marcinkute R, Woodward ER, Gandhi A, et al. Uptake and efficacy of bilateral risk reducing surgery in unaffected female BRCA1 and BRCA2 carriers. J Med Genet 2022;59:133–140.
- 46.↑
Parker WH, Feskanich D, Broder MS, et al. Long-term mortality associated with oophorectomy compared with ovarian conservation in the nurses’ health study. Obstet Gynecol 2013;121:709–716.
- 47.↑
Rivera CM, Grossardt BR, Rhodes DJ, et al. Increased cardiovascular mortality after early bilateral oophorectomy. Menopause 2009;16:15–23.
- 48.↑
Cancer Research UK. Breast cancer incidence (invasive) statistics. Accessed March 14, 2018. Available at: http://www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/breast-cancer/incidence-invasive#collapseOne
- 49.↑
UK Office for National Statistics. Cohort fertility: England and Wales. Accessed March 20, 2018. Available at: https://www.ons.gov.uk/peoplepopulationandcommunity/birthsdeathsandmarriages/conceptionandfertilityrates/datasets/cohortfertilityenglandandwales
- 50.↑
National Center for Health Statistics. Cohort fertility tables. Accessed November 20, 2018. Available at: https://www.cdc.gov/nchs/nvss/cohort_fertility_tables.htm
- 51.↑
Manchanda R, Burnell M, Loggenberg K, et al. Cluster-randomised non-inferiority trial comparing DVD-assisted and traditional genetic counselling in systematic population testing for BRCA1/2 mutations. J Med Genet 2016;53:472–480.
- 52.↑
Schwartz MD, Valdimarsdottir HB, Peshkin BN, et al. Randomized noninferiority trial of telephone versus in-person genetic counseling for hereditary breast and ovarian cancer. J Clin Oncol 2014;32:618–626.
- 53.↑
Curtis L, Burns A, eds. Unit costs of health and social care 2016. Accessed January 8, 2018. Available at: https://www.pssru.ac.uk/project-pages/unit-costs/unit-costs-2016/
- 54.↑
NHS Improvement. Archived reference costs. 2016/17 reference costs. Accessed January 8, 2018. Available at: https://webarchive.nationalarchives.gov.uk/ukgwa/20200501111106/https://improvement.nhs.uk/resources/reference-costs/
- 55.↑
Curtis L. Unit costs of health and social care 2011. Accessed January 8, 2018. Available at: https://www.pssru.ac.uk/project-pages/unit-costs/unit-costs-2011/
- 56.↑
Guy H, Walder L, Fisher M. Cost-effectiveness of niraparib versus routine surveillance, olaparib and rucaparib for the maintenance treatment of patients with ovarian cancer in the United States. Pharmacoeconomics 2019;37:391–405.
- 57.↑
National Institute for Health and Care Excellence. NICE health technology evaluations: the manual. Accessed November 15, 2023. Available at: https://www.nice.org.uk/process/pmg36/chapter/introduction-to-health-technology-evaluation
- 58.↑
Evans DG, Lalloo F, Ashcroft L, et al. Uptake of risk-reducing surgery in unaffected women at high risk of breast and ovarian cancer is risk, age, and time dependent. Cancer Epidemiol Biomarkers Prev 2009;18:2318–2324.
- 59.↑
Allemani C, Matsuda T, Di Carlo V, et al. Global surveillance of trends in cancer survival 2000–14 (CONCORD-3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet 2018;391:1023–1075.
- 60.↑
Bordeleau L, Panchal S, Goodwin P. Prognosis of BRCA-associated breast cancer: a summary of evidence. Breast Cancer Res Treat 2010;119:13–24.
- 61.↑
Rennert G, Bisland-Naggan S, Barnett-Griness O, et al. Clinical outcomes of breast cancer in carriers of BRCA1 and BRCA2 mutations. N Engl J Med 2007;357:115–123.
- 62.↑
Hammerschmidt T, Goertz A, Wagenpfeil S, et al. Validation of health economic models: the example of EVITA. Value Health 2003;6:551–559.
- 63.↑
National Institute for Health and Care Excellence. Guide to the methods of technology appraisal 2013. Accessed March 31, 2019. Available at: https://www.nice.org.uk/process/pmg9/resources/guide-to-the-methods-of-technology-appraisal-2013-pdf-2007975843781
- 64.↑
Neumann PJ, Cohen JT, Weinstein MC. Updating cost-effectiveness—the curious resilience of the $50,000-per-QALY threshold. N Engl J Med 2014;371:796–797.
- 65.↑
Morgan RD, Burghel GJ, Flaum N, et al. BRCA1/2 in non-mucinous epithelial ovarian cancer: tumour with or without germline testing? Br J Cancer 2022;127:163–167.
- 66.↑
Briggs A. Probabilistic analysis of cost-effectiveness models: statistical representation of parameter uncertainty. Value Health 2005;8:1–2.
- 67.↑
Yang X, Leslie G, Doroszuk A, et al. Cancer risks associated with germline PALB2 pathogenic variants: an international study of 524 families. J Clin Oncol 2020;38:674–685.
- 68.↑
Pal T, Akbari MR, Sun P, et al. Frequency of mutations in mismatch repair genes in a population-based study of women with ovarian cancer. Br J Cancer 2012;107:1783–1790.
- 69.↑
Song H, Cicek MS, Dicks E, et al. The contribution of deleterious germline mutations in BRCA1, BRCA2 and the mismatch repair genes to ovarian cancer in the population. Hum Mol Genet 2014;23:4703–4709.
- 70.↑
Minion LE, Dolinsky JS, Chase DM, et al. Hereditary predisposition to ovarian cancer, looking beyond BRCA1/BRCA2. Gynecol Oncol 2015;137:86–92.
- 71.↑
Kwon JS, Tinker AV, Hanley GE, et al. BRCA mutation testing for first-degree relatives of women with high-grade serous ovarian cancer. Gynecol Oncol 2019;152:459–464.
- 72.↑
Gao W, Muston D, Monberg M, et al. A critical appraisal and recommendations for cost-effectiveness studies of poly(ADP-ribose) polymerase inhibitors in advanced ovarian cancer. Pharmacoeconomics 2020;38:1201–1218.
- 73.↑
Dottino JA, Moss HA, Lu KH, et al. U.S. Food and Drug Administration-approved poly (ADP-ribose) polymerase inhibitor maintenance therapy for recurrent ovarian cancer: a cost-effectiveness analysis. Obstet Gynecol 2019;133:795–802.
- 74.↑
Zhong L, Tran AT, Tomasino T, et al. Cost-effectiveness of niraparib and olaparib as maintenance therapy for patients with platinum-sensitive recurrent ovarian cancer. J Manag Care Spec Pharm 2018;24:1219–1228.
- 75.↑
Sundar S, Manchanda R, Gourley C, et al. British Gynaecological Cancer Society/British Association of Gynaecological Pathology consensus for germline and tumour testing for BRCA1/2 variants in ovarian cancer in the United Kingdom. Accessed September 13, 2020. Available at: https://www.bgcs.org.uk/wp-content/uploads/2020/09/BGCS-BAGP-070920-final-v1.pdf
- 76.↑
Penn CA, Wong MS, Walsh CS. Cost-effectiveness of maintenance therapy based on molecular classification following treatment of primary epithelial ovarian cancer in the United States. JAMA Netw Open 2020;3:e2028620.
- 77.↑
Boussios S, Rassy E, Moschetta M, et al. BRCA mutations in ovarian and prostate cancer: bench to bedside. Cancers (Basel) 2022;14:3888.
- 78.↑
American Society of Clinical Oncology. American Society of Clinical Oncology position statement on addressing the affordability of cancer drugs. J Oncol Pract 2018;14:187–192.
- 79.↑
WHO. Pricing of cancer medicines and its impact. Accessed November 15, 2023. Available at: https://iris.who.int/bitstream/handle/10665/277190/9789241515115-eng.pdf?ua=1