1
|
Torr B, Jones C, Kavanaugh G, Hamill M, Allen S, Choi S, Garrett A, Valganon-Petrizan M, MacMahon S, Yuan L, Way R, Harder H, Gold R, Taylor A, Gabe R, Lucassen A, Manchanda R, Fallowfield L, Jenkins V, Gandhi A, Evans DG, George A, Hubank M, Kemp Z, Bremner S, Turnbull C. BRCA-DIRECT digital pathway for diagnostic germline genetic testing within a UK breast oncology setting: a randomised, non-inferiority trial. Br J Cancer 2024; 131:1506-1515. [PMID: 39349619 PMCID: PMC11519354 DOI: 10.1038/s41416-024-02832-2] [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: 05/10/2024] [Revised: 08/13/2024] [Accepted: 08/19/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Genetic testing to identify germline high-risk pathogenic variants in breast cancer susceptibility genes is increasingly part of the breast cancer diagnostic pathway. Novel patient-centred pathways may offer opportunity to expand capacity and reduce turnaround time. METHODS We recruited 1140 women with unselected breast cancer to undergo germline genetic testing through the BRCA-DIRECT pathway (which includes a digital platform, postal saliva sampling and a genetic counsellor telephone helpline). Ahead of consenting to the test, participants were randomised to receive information about genetic testing digitally (569/1140, 49.9%) or via a pre-test genetic counselling consultation (571/1140, 50.1%). RESULTS 1001 (87.8%) participants progressed to receive their pre-test information and consented to testing. The primary outcome, uptake of genetic testing, was higher amongst participants randomised to receive digital information compared with those randomised to a pre-test genetic counselling consultation (90.8% (95% CI: 88.5% to 93.1%) vs 84.7% (95% CI: 81.8% to 87.6%), p = 0.002, adjusted for participant age and site). Non-inferiority was observed in relation to patient knowledge, anxiety, and satisfaction. CONCLUSIONS Findings demonstrate that standardised, digital information offers a non-inferior alternative to conventional genetic counselling, and an end-to-end patient-centred, digital pathway (supported by genetic counselling hotline) could feasibly be implemented into breast oncology settings. CLINICAL TRIAL REGISTRATION The study is registered with, and protocol available on, ClinicalTrials.gov (NCT04842799).
Collapse
Affiliation(s)
- Bethany Torr
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
| | - Christopher Jones
- Brighton and Sussex Clinical Trials Unit, Brighton and Sussex Medical School, Brighton, UK
| | - Grace Kavanaugh
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
| | - Monica Hamill
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
| | - Sophie Allen
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
| | - Subin Choi
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
| | - Alice Garrett
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
- Department of Clinical Genetics, St Georges Hospital NHS Trust, London, UK
| | - Mikel Valganon-Petrizan
- Clinical Genomics Department, Centre for Molecular Pathology, NIHR Cancer Biomedical Research Centre, Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Suzanne MacMahon
- Clinical Genomics Department, Centre for Molecular Pathology, NIHR Cancer Biomedical Research Centre, Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Lina Yuan
- Clinical Genomics Department, Centre for Molecular Pathology, NIHR Cancer Biomedical Research Centre, Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Rosalind Way
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
| | - Helena Harder
- Sussex Health Outcomes Research and Education in Cancer (SHORE-C), Brighton and Sussex Medical School, Brighton, UK
| | | | - Amy Taylor
- Clinical Genetics, East Anglian Medical Genetics Service, Cambridge, UK
| | - Rhian Gabe
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Anneke Lucassen
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ranjit Manchanda
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
- Department of Gynaecological Oncology, Barts Health NHS Trust, London, UK
- Department of Health Services Research, Faculty of Public Health & Policy, London School of Hygiene and Tropical Medicine, London, UK
| | - Lesley Fallowfield
- Sussex Health Outcomes Research and Education in Cancer (SHORE-C), Brighton and Sussex Medical School, Brighton, UK
| | - Valerie Jenkins
- Sussex Health Outcomes Research and Education in Cancer (SHORE-C), Brighton and Sussex Medical School, Brighton, UK
| | - Ashu Gandhi
- School of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Prevent Breast Cancer Centre, Wythenshawe Hospital Manchester Universities Foundation Trust, Manchester, UK
| | - D Gareth Evans
- Nightingale and Genesis Breast Cancer Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
- Division of Evolution, Infection, and Genomic Sciences, The University of Manchester, Manchester, UK
| | - Angela George
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
- Cancer Genetics Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Michael Hubank
- Clinical Genomics Department, Centre for Molecular Pathology, NIHR Cancer Biomedical Research Centre, Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Zoe Kemp
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
- Cancer Genetics Unit, Royal Marsden NHS Foundation Trust, London, UK
- Breast Oncology Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Stephen Bremner
- Brighton and Sussex Clinical Trials Unit, Brighton and Sussex Medical School, Brighton, UK
| | - Clare Turnbull
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK.
- Cancer Genetics Unit, Royal Marsden NHS Foundation Trust, London, UK.
| |
Collapse
|
2
|
Abdel-Razeq H, Sharaf B, Tamimi F, Hani HB, Alsmadi O, Khalil H, Abunasser M, Edaily S, Mansour A. Establishment of a clinical cancer genetics program for breast cancer in a resource-limited country; challenges and opportunities. Front Oncol 2024; 14:1431985. [PMID: 39507757 PMCID: PMC11537866 DOI: 10.3389/fonc.2024.1431985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Breast cancer is the most common cancer among women worldwide, and its incidence rate is still increasing, especially among younger women. Nationally, it constitutes one-fifth of all cancer cases and almost 40% of all female cancers. With a median age of 51 years, breast cancer is diagnosed at least a decade earlier, and at more advanced stages compared to Western societies. Hereditary cancers account for 10% or more of all cancer burden worldwide. With expanded indications, increased number of genes tested, and significant decline in cost of testing, such proportion will probably increase. Individuals with pathogenic variants of BRCA1 and BRCA2 are at higher risk of breast, ovarian, pancreatic and many other cancers. Over the past two decades, several highly penetrant cancer-susceptibility genes were identified across almost all tumor sites, thus increasing the need for comprehensive cancer genetic programs that address the testing process, counselling patients and at-risk family members, and then deal with all testing results and its consequences. In addition to its important role in preventing more cancers in index patients themselves and among their close relatives, identification of pathogenic or likely pathogenic variants, mostly in BRCA1 or BRCA2, may inform therapeutic decisions in common cancers including breast, ovarian, prostate and pancreatic cancers. In this manuscript, we describe the experience of a comprehensive cancer center, in a resource-limited country in establishing a comprehensive clinical cancer genetics program that can serve as an example for others who share similar demographic and financial restrains.
Collapse
Affiliation(s)
- Hikmat Abdel-Razeq
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Baha Sharaf
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Faris Tamimi
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Hira Bani Hani
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Osama Alsmadi
- Department of Cell Therapy and Applied Genomics, King Hussein Cancer Center, Amman, Jordan
| | - Hanan Khalil
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Mahmoud Abunasser
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Sarah Edaily
- Department of Internal Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Asem Mansour
- Department of Radiology, King Hussein Cancer Center, Amman, Jordan
| |
Collapse
|
3
|
Dubsky P, Jackisch C, Im SA, Hunt KK, Li CF, Unger S, Paluch-Shimon S. BRCA genetic testing and counseling in breast cancer: how do we meet our patients' needs? NPJ Breast Cancer 2024; 10:77. [PMID: 39237557 PMCID: PMC11377442 DOI: 10.1038/s41523-024-00686-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/19/2024] [Indexed: 09/07/2024] Open
Abstract
BRCA1 and BRCA2 are tumor suppressor genes that have been linked to inherited susceptibility of breast cancer. Germline BRCA1/2 pathogenic or likely pathogenic variants (gBRCAm) are clinically relevant for treatment selection in breast cancer because they confer sensitivity to poly(ADP-ribose) polymerase (PARP) inhibitors. BRCA1/2 mutation status may also impact decisions on other systemic therapies, risk-reducing measures, and choice of surgery. Consequently, demand for gBRCAm testing has increased. Several barriers to genetic testing exist, including limited access to testing facilities, trained counselors, and psychosocial support, as well as the financial burden of testing. Here, we describe current implications of gBRCAm testing for patients with breast cancer, summarize current approaches to gBRCAm testing, provide potential solutions to support wider adoption of mainstreaming testing practices, and consider future directions of testing.
Collapse
Affiliation(s)
- Peter Dubsky
- Breast and Tumor Center, Hirslanden Klinik St. Anna, Lucerne, Switzerland.
- University of Lucerne, Faculty of Health Sciences and Medicine, Lucerne, Switzerland.
| | - Christian Jackisch
- Department of Obstetrics and Gynecology, Breast and Gynecologic Cancer Center, Sana Klinikum Offenbach, Offenbach, Germany
| | - Seock-Ah Im
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University, Seoul, Republic of Korea
| | | | - Chien-Feng Li
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | | | - Shani Paluch-Shimon
- Hadassah University Hospital & Faculty of Medicine, Hebrew University, Jerusalem, Israel
| |
Collapse
|
4
|
Ghareeb GA, Nass ZA, Abu-Grain S, Alnaji A, Almohanna H, Nasser HAS, Al Shahrani S. Genetic Testing Among Breast Cancer Patients in the Eastern Region of Saudi Arabia: Single-Center Experience. J Epidemiol Glob Health 2024; 14:1351-1357. [PMID: 39256315 PMCID: PMC11442735 DOI: 10.1007/s44197-024-00296-4] [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: 11/25/2023] [Accepted: 09/01/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Genetic testing for persons with a heightened likelihood of harboring a germline mutation permits early identification and appropriate management. This study aimed to identify the proportion of breast cancer (BC) patients who were offered genetic testing and the prevalence of BRCA mutations among them. Additionally, we assessed the demographic and clinical features of BC patients in the Eastern Region of Saudi Arabia. MATERIALS AND METHODS Data from 2535 patients with BC were retrieved from the registry between 2017 and 2021. The data were analyzed and presented using univariate and bivariate statistics. Odds ratios and 95% confidence intervals using logistic regression analysis were computed to identify the predictors of BRCA testing. RESULTS Patients with BC ranged in age from 18 to 103 years, and the mean age was 49.60 ± 12.14 years. BC was detected in men in 29 (1.1%) cases. Among diagnosed patients with BC, a total of 96 (3.7%) patients underwent testing for BRCA gene mutations. Of them, 36 (37.5%) patients had a BRCA gene mutation. The likelihood of undergoing BRCA testing was higher for those who were diagnosed with the condition before the age of 50, patients who were referred from private institutions, and patients with a history of previously diagnosed cancer. The likelihood of conducting BRCA testing was significantly lower among those with distant metastases. CONCLUSION The proportion of BRCA testing among BC patients was found to be relatively low. The development of a cost-effective, locally developed risk assessment tool that incorporates genetic counseling and testing for those with a familial predisposition to BC is imperative.
Collapse
Affiliation(s)
- Ghadeer Al Ghareeb
- Qatif Health Network, Eastern Health Cluster, Dammam, Kingdom of Saudi Arabia.
| | - Zainab Al Nass
- Qatif Health Network, Eastern Health Cluster, Dammam, Kingdom of Saudi Arabia
| | - Salma Abu-Grain
- Qatif Health Network, Eastern Health Cluster, Dammam, Kingdom of Saudi Arabia
| | - Alia Alnaji
- Qatif Health Network, Eastern Health Cluster, Dammam, Kingdom of Saudi Arabia
| | - Hani Almohanna
- Research Centre- KKMC, King Fahad Specialist Hospital-Dammam, Eastern Health Cluster, Dammam, Kingdom of Saudi Arabia
| | | | - Saad Al Shahrani
- King Fahad Specialist Hospital- Dammam, Eastern Health Cluster, Dammam, Kingdom of Saudi Arabia
| |
Collapse
|
5
|
Dioun SM, Perez LR, Prabhu M, Brewer JT, Ahsan MD, Hou JY, Sharaf RN, Wright JD, Frey MK. Cost-effectiveness of BRCA1 testing at time of obstetrical prenatal carrier screening for cancer prevention. Am J Obstet Gynecol 2024; 231:330.e1-330.e14. [PMID: 38621481 DOI: 10.1016/j.ajog.2024.04.014] [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: 01/10/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Improved technologies paired with an increase in access to genetic testing have led to the availability of expanded carrier screening evaluating hundreds of disorders. Currently, most autosomal dominant mutations, such as BRCA1, are not included in expanded carrier assays. Screening pregnant or preconception reproductive-aged women for BRCA1 may present a unique opportunity to perform population-based screening for patients at a time when precancer screening, chemoprevention, and/or risk-reducing surgery may be beneficial. OBJECTIVE This study aimed to inform clinical decision-making as to whether the universal incorporation of BRCA1 testing at the time of obstetrical prenatal carrier screening is cost-effective. STUDY DESIGN A decision analysis and Markov model was created. The initial decision point in the model was BRCA1 testing at the time of expanded carrier screening. Model probabilities, cost, and utility values were derived from published literature. For BRCA1-positive patients, the model simulated breast cancer screening and risk-reducing surgical interventions. A cycle length of 1 year and a time horizon of 47 years were used to simulate the lifespan of patients. The setting was obstetrical clinics in the United States, and the participants were a theoretical cohort of 1,429,074 pregnant patients who annually underwent expanded carrier screening. RESULTS Among our cohort, BRCA1 testing resulted in the identification of an additional 3716 BRCA1-positive patients, the prevention of 1394 breast and ovarian cancer cases, and 1084 fewer deaths. BRCA1 testing was a cost-effective strategy compared with no BRCA1 testing with an incremental cost-effectiveness ratio of $86,001 per quality-adjusted life years. In a 1-way sensitivity analysis, we varied the prevalence of BRCA1 in the population from 0.00% to 20.00% and found that BRCA1 testing continued to be the cost-effective strategy until the prevalence rate was reduced to 0.16%. Multiple additional sensitivity analyses did not substantially affect the cost-effectiveness. CONCLUSION The addition of BRCA1 testing to obstetrical prenatal carrier screening is a cost-effective management strategy to identify at-risk women at a time when cancer screening and preventive strategies can be effective. Despite the burden of additional genetic counseling, prenatal care represents a unique opportunity to implement population-based genetic testing.
Collapse
Affiliation(s)
- Shayan M Dioun
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, NY; NewYork-Presbyterian Hospital, New York, NY.
| | | | - Malavika Prabhu
- Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | | | | | - June Y Hou
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, NY; NewYork-Presbyterian Hospital, New York, NY
| | | | - Jason D Wright
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, NY; NewYork-Presbyterian Hospital, New York, NY
| | - Melissa K Frey
- NewYork-Presbyterian Hospital, New York, NY; Weill Cornell Medicine, New York, NY
| |
Collapse
|
6
|
Ehman M, Punian J, Weymann D, Regier DA. Next-generation sequencing in oncology: challenges in economic evaluations. Expert Rev Pharmacoecon Outcomes Res 2024:1-18. [PMID: 39096135 DOI: 10.1080/14737167.2024.2388814] [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: 06/20/2024] [Revised: 07/19/2024] [Accepted: 08/01/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Next-generation sequencing (NGS) identifies genetic variants to inform personalized treatment plans. Insufficient evidence of cost-effectiveness impedes the integration of NGS into routine cancer care. The complexity of personalized treatment challenges conventional economic evaluation. Clearly delineating challenges informs future cost-effectiveness analyses to better value and contextualize health, preference-, and equity-based outcomes. AREAS COVERED We conducted a scoping review to characterize the applied methods and outcomes of economic evaluations of NGS in oncology and identify existing challenges. We included 27 articles published since 2016 from a search of PubMed, Embase, and Web of Science. Identified challenges included defining the evaluative scope, managing evidentiary limitations including lack of causal evidence, incorporating preference-based utility, and assessing distributional and equity-based impacts. These challenges reflect the difficulty of generating high-quality clinical effectiveness and real-world evidence (RWE) for NGS-guided interventions. EXPERT OPINION Adapting methodological approaches and developing life-cycle health technology assessment (HTA) guidance using RWE is crucial for implementing NGS in oncology. Healthcare systems, decision-makers, and HTA organizations are facing a pivotal opportunity to adapt to an evolving clinical paradigm and create innovative regulatory and reimbursement processes that will enable more sustainable, equitable, and patient-oriented healthcare.
Collapse
Affiliation(s)
- Morgan Ehman
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
| | - Jesman Punian
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
| | - Deirdre Weymann
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
- Faculty of Health Sciences, Simon Fraser University, Vancouver, BC, Canada
| | - Dean A Regier
- Cancer Control Research, BC Cancer, Vancouver, BC, Canada
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
7
|
Basu S, Venkataramani AS, Schillinger D. The Risk Of Perpetuating Health Disparities Through Cost-Effectiveness Analyses. Health Aff (Millwood) 2024; 43:1165-1171. [PMID: 39102593 DOI: 10.1377/hlthaff.2023.01583] [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] [Indexed: 08/07/2024]
Abstract
Cost-effectiveness analyses are commonly used to inform health care and public health policy decisions. However, standard approaches may systematically disadvantage marginalized groups by incorporating assumptions of persisting health inequities. We examined how competing risks, baseline health care costs, and indirect costs can differentially affect cost-effectiveness analyses for racial and ethnic minority populations. We illustrate that these structural factors can reduce estimated quality-adjusted life-years and cost savings for disadvantaged groups, making interventions focused on disadvantaged populations appear less cost-effective. For example, analyses of a sugar-sweetened beverage tax may estimate higher costs per quality-adjusted life-year gained for Black versus White populations because of differences in competing risks and insurance status that manifest in higher health care cost savings from averted disease among White people. To ensure that cost-effectiveness assessments do not perpetuate inequities, alternative approaches are needed that account for the impact of structural factors on different groups and that consider scenarios in which health inequities are reduced. Sensitivity analyses focusing on health equity could help advance interventions that disproportionately benefit disadvantaged communities.
Collapse
Affiliation(s)
- Sanjay Basu
- Sanjay Basu , Waymark, San Francisco, California
| | | | - Dean Schillinger
- Dean Schillinger, University of California San Francisco, San Francisco, California
| |
Collapse
|
8
|
Rios JD, Simbulan F, Reichman L, Caswell K, Tachdjian M, Malkin D, Cotton C, Nathan PC, Goudie C, Pechlivanoglou P. Cost-effectiveness of the McGill interactive pediatric oncogenetic guidelines in identifying Li-Fraumeni syndrome in female patients with osteosarcoma. Pediatr Blood Cancer 2024; 71:e31077. [PMID: 38783403 DOI: 10.1002/pbc.31077] [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: 11/08/2023] [Revised: 03/20/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Li-Fraumeni syndrome (LFS) is a penetrant cancer predisposition syndrome (CPS) associated with the development of many tumor types in young people including osteosarcoma and breast cancer (BC). The McGill Interactive Pediatric OncoGenetic Guidelines (MIPOGG) decision-support tool provides a standardized approach to identify patients at risk of CPSs. METHODS We conducted a cost-utility analysis, from the healthcare payer perspective, to compare MIPOGG-guided, physician-guided, and universal genetic testing strategies to detect LFS in female patients diagnosed at an age of less than 18 years with osteosarcoma. We developed a decision tree and discrete-event simulation model to simulate the clinical and cost outcomes of the three genetic referral strategies on a cohort of female children diagnosed with osteosarcoma, especially focused on BC as subsequent cancer. Outcomes included BC incidence, quality-adjusted life-years (QALYs), healthcare costs, and incremental cost-utility ratios (ICURs). We conducted probabilistic and scenario analyses to assess the uncertainty surrounding model parameters. RESULTS Compared to the physician-guided testing, the MIPOGG-guided strategy was marginally more expensive by $105 (-$516; $743), but slightly more effective by 0.003 (-0.04; 0.045) QALYs. Compared to MIPOGG, the universal testing strategy was $1333 ($732; $1953) more costly and associated with 0.011 (-0.043; 0.064) additional QALYs. The ICUR for the MIPOGG strategy was $33,947/QALY when compared to the physician strategy; the ICUR for universal testing strategy was $118,631/QALY when compared to the MIPOGG strategy. DISCUSSION This study provides evidence for clinical and policy decision-making on the cost-effectiveness of genetic referral strategies to identify LFS in the setting of osteosarcoma. MIPOGG-guided strategy was most likely to be cost-effective at a willingness-to-pay threshold value of $50,000/QALY.
Collapse
Affiliation(s)
- Juan David Rios
- Child Health Evaluative Sciences, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Frances Simbulan
- Child Health Evaluative Sciences, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Lara Reichman
- Child Health and Human Development, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Kimberly Caswell
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Melissa Tachdjian
- Child Health and Human Development, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - David Malkin
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Cecilia Cotton
- Department of Statistics and Actuarial Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Paul C Nathan
- Child Health Evaluative Sciences, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Catherine Goudie
- Child Health and Human Development, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Pediatrics, Division of Hematology-Oncology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Petros Pechlivanoglou
- Child Health Evaluative Sciences, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Ashton-Prolla P, Achatz MIW, Moreira MAM, Palmero EI, Soares DCDQ, Ferraz VEDF, Caires IQDS, Guindalini RSC, Gifoni ACLVC. Importance of genetic cancer risk assessment as a strategy to stratify risk and provide precision prevention in high-risk patients and families. REVISTA DA ASSOCIACAO MEDICA BRASILEIRA (1992) 2024; 70:e2024S117. [PMID: 38865537 PMCID: PMC11164255 DOI: 10.1590/1806-9282.2024s117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/31/2023] [Indexed: 06/14/2024]
Affiliation(s)
- Patricia Ashton-Prolla
- Hospital de Clínicas de Porto Alegre, Medical Genetics Service – Porto Alegre (RS), Brazil
- Universidade Federal do Rio Grande do Sul, Department of Genetics – Porto Alegre (RS), Brazil
| | | | | | - Edenir Inez Palmero
- Instituto Nacional de Câncer, Genetics Program – Rio de Janeiro (RJ), Brazil
| | - Diogo Cordeiro de Queiroz Soares
- Real Hospital Português, Oncogenetics Service – Recife (PE), Brazil
- A.C. Camargo Cancer Center, Department of Oncogenetics – São Paulo (SP), Brazil
| | | | | | | | | |
Collapse
|
10
|
Chen S, Tse K, Lu Y, Chen S, Tian Y, Tan KT, Li C. Comprehensive genomic profiling and therapeutic implications for Taiwanese patients with treatment-naïve breast cancer. Cancer Med 2024; 13:e7384. [PMID: 38895905 PMCID: PMC11187859 DOI: 10.1002/cam4.7384] [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: 11/06/2023] [Revised: 03/29/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Breast cancer is a heterogeneous disease categorized based on molecular characteristics, including hormone receptor (HR) and human epidermal growth factor receptor 2 (HER2) expression levels. The emergence of profiling technology has revealed multiple driver genomic alterations within each breast cancer subtype, serving as biomarkers to predict treatment outcomes. This study aimed to explore the genomic landscape of breast cancer in the Taiwanese population through comprehensive genomic profiling (CGP) and identify diagnostic and predictive biomarkers. METHODS Targeted next-generation sequencing-based CGP was performed on 116 archived Taiwanese breast cancer specimens, assessing genomic alterations (GAs), including single nucleotide variants, copy number variants, fusion genes, tumor mutation burden (TMB), and microsatellite instability (MSI) status. Predictive variants for FDA-approved therapies were evaluated within each subtype. RESULTS In the cohort, frequent mutations included PIK3CA (39.7%), TP53 (36.2%), KMT2C (9.5%), GATA3 (8.6%), and SF3B1 (6.9%). All subtypes had low TMB, with no MSI-H tumors. Among HR + HER2- patients, 42% (27/65) harbored activating PIK3CA mutations, implying potential sensitivity to PI3K inhibitors and resistance to endocrine therapies. HR + HER2- patients exhibited intrinsic hormonal resistance via FGFR1 gene gain/amplification (15%), exclusive of PI3K/AKT pathway alterations. Aberrations in the PI3K/AKT/mTOR and FGFR pathways were implicated in chemoresistance, with a 52.9% involvement in triple-negative breast cancer. In HER2+ tumors, 50% harbored GAs potentially conferring resistance to anti-HER2 therapies, including PIK3CA mutations (32%), MAP3K1 (2.9%), NF1 (2.9%), and copy number gain/amplification of FGFR1 (18%), FGFR3 (2.9%), EGFR (2.9%), and AKT2 (2.9%). CONCLUSION This study presents CGP findings for treatment-naïve Taiwanese breast cancer, emphasizing its value in routine breast cancer management, disease classification, and treatment selection.
Collapse
Affiliation(s)
- Shang‐Hung Chen
- National Institute of Cancer Research, National Health Research InstitutesTainanTaiwan
- Department of OncologyNational Cheng Kung University Hospital, College of Medicine, National Cheng Kung UniversityTainanTaiwan
| | | | | | | | - Yu‐Feng Tian
- Division of Colorectal Surgery, Department of SurgeryChi Mei Medical CenterTainanTaiwan
- Department of Health and NutritionChia‐Nan University of Pharmacy and ScienceTainanTaiwan
| | - Kien Thiam Tan
- ACT Genomics, Co. Ltd.TaipeiTaiwan
- Anbogen Therapeutics, Inc.TaipeiTaiwan
| | - Chien‐Feng Li
- National Institute of Cancer Research, National Health Research InstitutesTainanTaiwan
- Department of Medical ResearchChi Mei Medical CenterTainanTaiwan
- Institute of Precision MedicineNational Sun Yat‐Sen UniversityKaohsiungTaiwan
- Department of Clinical Pathology and Laboratory MedicineChi Mei Medical CenterTainanTaiwan
- Trans‐omic Laboratory for Precision MedicineChi Mei Medical CenterTainanTaiwan
| |
Collapse
|
11
|
Yang DW, Miller JA, Xue WQ, Tang M, Lei L, Zheng Y, Diao H, Wang TM, Liao Y, Wu YX, Zheng XH, Zhou T, Li XZ, Zhang PF, Chen XY, Yu X, Li F, Ji M, Sun Y, He YQ, Jia WH. Polygenic risk-stratified screening for nasopharyngeal carcinoma in high-risk endemic areas of China: a cost-effectiveness study. Front Public Health 2024; 12:1375533. [PMID: 38756891 PMCID: PMC11097958 DOI: 10.3389/fpubh.2024.1375533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
Background Nasopharyngeal carcinoma (NPC) has an extremely high incidence rate in Southern China, resulting in a severe disease burden for the local population. Current EBV serologic screening is limited by false positives, and there is opportunity to integrate polygenic risk scores for personalized screening which may enhance cost-effectiveness and resource utilization. Methods A Markov model was developed based on epidemiological and genetic data specific to endemic areas of China, and further compared polygenic risk-stratified screening [subjects with a 10-year absolute risk (AR) greater than a threshold risk underwent EBV serological screening] to age-based screening (EBV serological screening for all subjects). For each initial screening age (30-34, 35-39, 40-44, 45-49, 50-54, 55-59, 60-64, and 65-69 years), a modeled cohort of 100,000 participants was screened until age 69, and then followed until age 79. Results Among subjects aged 30 to 54 years, polygenic risk-stratified screening strategies were more cost-effective than age-based screening strategies, and almost comprised the cost-effectiveness efficiency frontier. For men, screening strategies with a 1-year frequency and a 10-year absolute risk (AR) threshold of 0.7% or higher were cost-effective, with an incremental cost-effectiveness ratio (ICER) below the willingness to pay (¥203,810, twice the local per capita GDP). Specifically, the strategies with a 10-year AR threshold of 0.7% or 0.8% are the most cost-effective strategies, with an ICER ranging from ¥159,752 to ¥201,738 compared to lower-cost non-dominated strategies on the cost-effectiveness frontiers. The optimal strategies have a higher probability (29.4-35.8%) of being cost-effective compared to other strategies on the frontier. Additionally, they reduce the need for nasopharyngoscopies by 5.1-27.7% compared to optimal age-based strategies. Likewise, for women aged 30-54 years, the optimal strategy with a 0.3% threshold showed similar results. Among subjects aged 55 to 69 years, age-based screening strategies were more cost-effective for men, while no screening may be preferred for women. Conclusion Our economic evaluation found that the polygenic risk-stratified screening could improve the cost-effectiveness among individuals aged 30-54, providing valuable guidance for NPC prevention and control policies in endemic areas of China.
Collapse
Affiliation(s)
- Da-Wei Yang
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jacob A. Miller
- Department of Radiation Oncology, Cleveland Clinic, Cleveland, OH, United States
| | - Wen-Qiong Xue
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | | | - Lin Lei
- Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Yuming Zheng
- Wuzhou Red Cross Hospital, Wuzhou, Guangxi, China
| | - Hua Diao
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Tong-Min Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ying Liao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan-Xia Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao-Hui Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ting Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xi-Zhao Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Pei-Fen Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xue-Yin Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xia Yu
- Cancer Research Institute of Zhongshan City, Zhongshan Hospital of Sun Yat-sen University, Zhongshan, China
| | - Fugui Li
- Cancer Research Institute of Zhongshan City, Zhongshan Hospital of Sun Yat-sen University, Zhongshan, China
| | - Mingfang Ji
- Cancer Research Institute of Zhongshan City, Zhongshan Hospital of Sun Yat-sen University, Zhongshan, China
| | - Ying Sun
- Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yong-Qiao He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wei-Hua Jia
- School of Public Health, Sun Yat-Sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
12
|
Banerjee S. Cost-Effectiveness and the Economics of Genomic Testing and Molecularly Matched Therapies. Surg Oncol Clin N Am 2024; 33:231-242. [PMID: 38401907 DOI: 10.1016/j.soc.2023.12.010] [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: 02/26/2024]
Abstract
Cost-effectiveness analysis of precision oncology can help guide value-driven care. Next-generation sequencing is increasingly cost-efficient over single gene testing because diagnostic algorithms require multiple individual gene tests to determine biomarker status. Matched targeted therapy is often not cost-effective due to the high cost associated with drug treatment. However, genomic profiling can promote cost-effective care by identifying patients who are unlikely to benefit from therapy. Additional applications of genomic profiling such as universal testing for hereditary cancer syndromes and germline testing in patients with cancer may represent cost-effective approaches compared with traditional history-based diagnostic methods.
Collapse
Affiliation(s)
- Sudeep Banerjee
- Division of Colorectal Surgery, Department of General Surgery, Kaiser Permanente San Jose Medical Center, Kaiser Permanente Northern California, 280 Hospital Parkway, Building B, San Jose, CA 95119, USA.
| |
Collapse
|
13
|
Sideris M, Menon U, Manchanda R. Screening and prevention of ovarian cancer. Med J Aust 2024; 220:264-274. [PMID: 38353066 DOI: 10.5694/mja2.52227] [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: 02/27/2023] [Accepted: 12/11/2023] [Indexed: 03/07/2024]
Abstract
Ovarian cancer remains the most lethal gynaecological malignancy with 314 000 cases and 207 000 deaths annually worldwide. Ovarian cancer cases and deaths are predicted to increase in Australia by 42% and 55% respectively by 2040. Earlier detection and significant downstaging of ovarian cancer have been demonstrated with multimodal screening in the largest randomised controlled trial of ovarian cancer screening in women at average population risk. However, none of the randomised trials have demonstrated a mortality benefit. Therefore, ovarian cancer screening is not currently recommended in women at average population risk. More frequent surveillance for ovarian cancer every three to four months in women at high risk has shown good performance characteristics and significant downstaging, but there is no available information on a survival benefit. Population testing offers an emerging novel strategy to identify women at high risk who can benefit from ovarian cancer prevention. Novel multicancer early detection biomarker, longitudinal multiple marker strategies, and new biomarkers are being investigated and evaluated for ovarian cancer screening. Risk-reducing salpingo-oophorectomy (RRSO) decreases ovarian cancer incidence and mortality and is recommended for women at over a 4-5% lifetime risk of ovarian cancer. Pre-menopausal women without contraindications to hormone replacement therapy (HRT) undergoing RRSO should be offered HRT until 51 years of age to minimise the detrimental consequences of premature menopause. Currently risk-reducing early salpingectomy and delayed oophorectomy (RRESDO) should only be offered to women at increased risk of ovarian cancer within the context of a research trial. Pre-menopausal early salpingectomy is associated with fewer menopausal symptoms and better sexual function than bilateral salpingo-oophorectomy. A Sectioning and Extensively Examining the Fimbria (SEE-FIM) protocol should be used for histopathological assessment in women at high risk of ovarian cancer who are undergoing surgical prevention. Opportunistic salpingectomy may be offered at routine gynaecological surgery to all women who have completed their family. Long term prospective opportunistic salpingectomy studies are needed to determine the effect size of ovarian cancer risk reduction and the impact on menopause.
Collapse
Affiliation(s)
- Michail Sideris
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Usha Menon
- Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Ranjit Manchanda
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
- Institute of Clinical Trials and Methodology, University College London, London, UK
- Barts Health NHS Trust, London, UK
- London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
14
|
Kansuttiviwat C, Lertwilaiwittaya P, Roothumnong E, Nakthong P, Dungort P, Meesamarnpong C, Tansa-Nga W, Pongsuktavorn K, Wiboonthanasarn S, Tititumjariya W, Phuphuripan N, Lertbussarakam C, Wattanarangsan J, Sritun J, Punuch K, Kammarabutr J, Mutirangura P, Thongnoppakhun W, Limwongse C, Pithukpakorn M. Germline mutations of 4567 patients with hereditary breast-ovarian cancer spectrum in Thailand. NPJ Genom Med 2024; 9:9. [PMID: 38355628 PMCID: PMC10866978 DOI: 10.1038/s41525-024-00400-4] [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: 09/15/2023] [Accepted: 01/29/2024] [Indexed: 02/16/2024] Open
Abstract
Multi-gene panel testing has led to the detection of pathogenic/likely pathogenic (P/LP) variants in many cancer susceptibility genes in patients with breast-ovarian cancer spectrum. However, the clinical and genomic data of Asian populations, including Thai cancer patients, was underrepresented, and the clinical significance of multi-gene panel testing in Thailand remains undetermined. In this study, we collected the clinical and genetic data from 4567 Thai patients with cancer in the hereditary breast-ovarian cancer (HBOC) spectrum who underwent multi-gene panel testing. Six hundred and ten individuals (13.4%) had germline P/LP variants. Detection rates of germline P/LP variants in breast, ovarian, pancreatic, and prostate cancer were 11.8%, 19.8%, 14.0%, and 7.1%, respectively. Non-BRCA gene mutations accounted for 35% of patients with germline P/LP variants. ATM was the most common non-BRCA gene mutation. Four hundred and thirty-two breast cancer patients with germline P/LP variants (80.4%) met the current NCCN genetic testing criteria. The most common indication was early-onset breast cancer. Ten patients harbored double pathogenic variants in this cohort. Our result showed that a significant proportion of non-BRCA P/LP variants were identified in patients with HBOC-related cancers. These findings support the benefit of multi-gene panel testing for inherited cancer susceptibility among Thai HBOC patients. Some modifications of the testing policy may be appropriate for implementation in diverse populations.
Collapse
Affiliation(s)
- Chalermkiat Kansuttiviwat
- Division of Medical Genetics, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pongtawat Lertwilaiwittaya
- Division of Medical Genetics, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ekkapong Roothumnong
- Division of Medical Genetics, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Panee Nakthong
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Peerawat Dungort
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chutima Meesamarnpong
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Warisara Tansa-Nga
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Khontawan Pongsuktavorn
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Supakit Wiboonthanasarn
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Warunya Tititumjariya
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nannipa Phuphuripan
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Jantanee Wattanarangsan
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jiraporn Sritun
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kittiporn Punuch
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jirayu Kammarabutr
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pornthira Mutirangura
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Wanna Thongnoppakhun
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chanin Limwongse
- Division of Medical Genetics, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Manop Pithukpakorn
- Division of Medical Genetics, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
15
|
Wei X, Sun L, Slade E, Fierheller CT, Oxley S, Kalra A, Sia J, Sideris M, McCluggage WG, Bromham N, Dworzynski K, Rosenthal AN, Brentnall A, Duffy S, Evans DG, Yang L, Legood R, Manchanda R. Cost-Effectiveness of Gene-Specific Prevention Strategies for Ovarian and Breast Cancer. JAMA Netw Open 2024; 7:e2355324. [PMID: 38334999 PMCID: PMC10858404 DOI: 10.1001/jamanetworkopen.2023.55324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/16/2023] [Indexed: 02/10/2024] Open
Abstract
Importance Pathogenic variants (PVs) in BRCA1, BRCA2, PALB2, RAD51C, RAD51D, and BRIP1 cancer susceptibility genes (CSGs) confer an increased ovarian cancer (OC) risk, with BRCA1, BRCA2, PALB2, RAD51C, and RAD51D PVs also conferring an elevated breast cancer (BC) risk. Risk-reducing surgery, medical prevention, and BC surveillance offer the opportunity to prevent cancers and deaths, but their cost-effectiveness for individual CSGs remains poorly addressed. Objective To estimate the cost-effectiveness of prevention strategies for OC and BC among individuals carrying PVs in the previously listed CSGs. Design, Setting, and Participants In this economic evaluation, a decision-analytic Markov model evaluated the cost-effectiveness of risk-reducing salpingo-oophorectomy (RRSO) and, where relevant, risk-reducing mastectomy (RRM) compared with nonsurgical interventions (including BC surveillance and medical prevention for increased BC risk) from December 1, 2022, to August 31, 2023. The analysis took a UK payer perspective with a lifetime horizon. The simulated cohort consisted of women aged 30 years who carried BRCA1, BRCA2, PALB2, RAD51C, RAD51D, or BRIP1 PVs. Appropriate sensitivity and scenario analyses were performed. Exposures CSG-specific interventions, including RRSO at age 35 to 50 years with or without BC surveillance and medical prevention (ie, tamoxifen or anastrozole) from age 30 or 40 years, RRM at age 30 to 40 years, both RRSO and RRM, BC surveillance and medical prevention, or no intervention. Main Outcomes and Measures The incremental cost-effectiveness ratio (ICER) was calculated as incremental cost per quality-adjusted life-year (QALY) gained. OC and BC cases and deaths were estimated. Results In the simulated cohort of women aged 30 years with no cancer, undergoing both RRSO and RRM was most cost-effective for individuals carrying BRCA1 (RRM at age 30 years; RRSO at age 35 years), BRCA2 (RRM at age 35 years; RRSO at age 40 years), and PALB2 (RRM at age 40 years; RRSO at age 45 years) PVs. The corresponding ICERs were -£1942/QALY (-$2680/QALY), -£89/QALY (-$123/QALY), and £2381/QALY ($3286/QALY), respectively. RRSO at age 45 years was cost-effective for RAD51C, RAD51D, and BRIP1 PV carriers compared with nonsurgical strategies. The corresponding ICERs were £962/QALY ($1328/QALY), £771/QALY ($1064/QALY), and £2355/QALY ($3250/QALY), respectively. The most cost-effective preventive strategy per 1000 PV carriers could prevent 923 OC and BC cases and 302 deaths among those carrying BRCA1; 686 OC and BC cases and 170 deaths for BRCA2; 464 OC and BC cases and 130 deaths for PALB2; 102 OC cases and 64 deaths for RAD51C; 118 OC cases and 76 deaths for RAD51D; and 55 OC cases and 37 deaths for BRIP1. Probabilistic sensitivity analysis indicated both RRSO and RRM were most cost-effective in 96.5%, 89.2%, and 84.8% of simulations for BRCA1, BRCA2, and PALB2 PVs, respectively, while RRSO was cost-effective in approximately 100% of simulations for RAD51C, RAD51D, and BRIP1 PVs. Conclusions and Relevance In this cost-effectiveness study, RRSO with or without RRM at varying optimal ages was cost-effective compared with nonsurgical strategies for individuals who carried BRCA1, BRCA2, PALB2, RAD51C, RAD51D, or BRIP1 PVs. These findings support personalizing risk-reducing surgery and guideline recommendations for individual CSG-specific OC and BC risk management.
Collapse
Affiliation(s)
- Xia Wei
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - Li Sun
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - Eric Slade
- National Institute for Health and Care Excellence, London, United Kingdom
| | - Caitlin T. Fierheller
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - Samuel Oxley
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
- Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London, United Kingdom
| | - Ashwin Kalra
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
- Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London, United Kingdom
| | - Jacqueline Sia
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
- Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London, United Kingdom
| | - Michail Sideris
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
- Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London, United Kingdom
| | - W. Glenn McCluggage
- Department of Pathology, Belfast Health & Social Care Trust, Royal Victoria Hospital, Belfast, United Kingdom
| | - Nathan Bromham
- National Institute for Health and Care Excellence, London, United Kingdom
| | | | - Adam N. Rosenthal
- Department of Gynaecology, University College London Hospitals NHS Foundation trust, London, United Kingdom
- Department of Women’s Cancer, UCL EGA Institute for Women’s Health, University College London, London, United Kingdom
| | - Adam Brentnall
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - Stephen Duffy
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - D. Gareth Evans
- Manchester Centre for Genomic Medicine, Division of Evolution, Infection and Genomic Sciences, University of Manchester, MAHSC, Manchester, United Kingdom
| | - Li Yang
- School of Public Health, Peking University, Beijing, China
| | - Rosa Legood
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
| | - Ranjit Manchanda
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Wolfson Institute of Population Health, Queen Mary University of London, London, United Kingdom
- Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London, United Kingdom
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials & Methodology, Faculty of Population Health Sciences, University College London, London, United Kingdom
| |
Collapse
|
16
|
Guo F, Adekanmbi V, Hsu CD, Berenson AB, Kuo YF, Shih YCT. Cost-Effectiveness of Population-Based Multigene Testing for Breast and Ovarian Cancer Prevention. JAMA Netw Open 2024; 7:e2356078. [PMID: 38353949 PMCID: PMC10867683 DOI: 10.1001/jamanetworkopen.2023.56078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 12/21/2023] [Indexed: 02/16/2024] Open
Abstract
Importance The current method of BRCA testing for breast and ovarian cancer prevention, which is based on family history, often fails to identify many carriers of pathogenic variants. Population-based genetic testing offers a transformative approach in cancer prevention by allowing for proactive identification of any high-risk individuals and enabling early interventions. Objective To assess the lifetime incremental effectiveness, costs, and cost-effectiveness of population-based multigene testing vs family history-based testing. Design, Setting, and Participants This economic evaluation used a microsimulation model to assess the cost-effectiveness of multigene testing (BRCA1, BRCA2, and PALB2) for all women aged 30 to 35 years compared with the current standard of care that is family history based. Carriers of pathogenic variants were offered interventions, such as magnetic resonance imaging with or without mammography, chemoprevention, or risk-reducing mastectomy and salpingo-oophorectomy, to reduce cancer risk. A total of 2000 simulations were run on 1 000 000 women, using a lifetime time horizon and payer perspective, and costs were adjusted to 2022 US dollars. This study was conducted from September 1, 2020, to December 15, 2023. Main Outcomes and Measures The main outcome measure was the incremental cost-effectiveness ratio (ICER), quantified as cost per quality-adjusted life-year (QALY) gained. Secondary outcomes included incremental cost, additional breast and ovarian cancer cases prevented, and excess deaths due to coronary heart disease (CHD). Results The study assessed 1 000 000 simulated women aged 30 to 35 years in the US. In the base case, population-based multigene testing was more cost-effective compared with family history-based testing, with an ICER of $55 548 per QALY (95% CI, $47 288-$65 850 per QALY). Population-based multigene testing would be able to prevent an additional 1338 cases of breast cancer and 663 cases of ovarian cancer, but it would also result in 69 cases of excess CHD and 10 excess CHD deaths per million women. The probabilistic sensitivity analyses show that the probability that population-based multigene testing is cost-effective was 100%. When the cost of the multigene test exceeded $825, population-based testing was no longer cost-effective (ICER, $100 005 per QALY; 95% CI, $87 601-$11 6323). Conclusions and Relevance In this economic analysis of population-based multigene testing, population-based testing was a more cost-effective strategy for the prevention of breast cancer and ovarian cancer when compared with the current family history-based testing strategy at the $100 000 per QALY willingness-to-pay threshold. These findings support the need for more comprehensive genetic testing strategies to identify pathogenic variant carriers and enable informed decision-making for personalized risk management.
Collapse
Affiliation(s)
- Fangjian Guo
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston
- Center for Interdisciplinary Research in Women’s Health, The University of Texas Medical Branch at Galveston, Galveston
| | - Victor Adekanmbi
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston
- Center for Interdisciplinary Research in Women’s Health, The University of Texas Medical Branch at Galveston, Galveston
| | - Christine D. Hsu
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston
- Center for Interdisciplinary Research in Women’s Health, The University of Texas Medical Branch at Galveston, Galveston
| | - Abbey B. Berenson
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston
- Center for Interdisciplinary Research in Women’s Health, The University of Texas Medical Branch at Galveston, Galveston
| | - Yong-Fang Kuo
- Center for Interdisciplinary Research in Women’s Health, The University of Texas Medical Branch at Galveston, Galveston
- Department of Biostatistics and Data Science, The University of Texas Medical Branch at Galveston, Galveston
- Office of Biostatistics, University of Texas Medical Branch at Galveston, Galveston
| | - Ya-Chen Tina Shih
- Program in Cancer Health Economics Research, Jonsson Comprehensive Cancer Center, and Department of Radiation Oncology, School of Medicine, University of California, Los Angeles
| |
Collapse
|
17
|
Liu S, Yan Y, Cui Z, Feng H, Zhong F, Liu Z, Li Y, Ou X, Li W. Relationship between PIWIL1 gene polymorphisms and epithelial ovarian cancer susceptibility among southern Chinese woman: a three-center case-control study. BMC Cancer 2023; 23:1149. [PMID: 38012622 PMCID: PMC10680212 DOI: 10.1186/s12885-023-11651-2] [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: 08/21/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023] Open
Abstract
OBJECTIVE To investigate the potential correlation between piwi-like RNA-mediated gene silencing 1 (PIWIL1) polymorphisms and susceptibility to epithelial ovarian cancer (EOC). METHODS A case-control study was conducted to evaluate the susceptibility of EOC using multinomial logistic regression analysis. The study analyzed the relationship between five functional single nucleotide polymorphisms (SNPs) in the PIWIL1 gene and EOC risk. Genotyping of 288 cases and 361 healthy samples from South China was identified using a TaqMan assay. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to estimate the relationship between the five selected SNPs and EOC susceptibility. RESULTS Among the five SNPs analyzed, the rs10848087 G > A and rs7957349 G > C variants significantly increased the susceptibility of EOC, rs10773771 C > T was associated with a decreased risk of EOC, while the rs35997018 and rs1106042 variants were not in Hardy-Weinberg equilibrium (p < 0.05). The rs10848087 G > A was significantly associated with increased risk of EOC in individuals with metastasis, FIGO stage I and III, low and high pathological grade, tumor numbers ≤ 3 and > 3, tumor size > 3 cm and ≤ 3 cm, pregnant more than 3 times, pre-menopausal status, and strong positive expression of ER (estrogen receptor), PR (progesterone receptor), PAX8 (paired-box 8), wild-type p53 (tumor protein 53), WT1 (Wilm's tumor gene), P16 (cyclin-dependent kinase inhibitor 2A). In addition, rs10848087 G > A enhanced the EOC risk of cases with negative/mild positive expression of wild p53 and Ki67, and with or without mutant p53 expression. The rs7957349 G > C variant was linked to an increased risk of EOC in subgroups with certain characteristics, including age equal or less than 53 years, metastasis, clinical stage I, low pathological grade, tumor number, tumor size, pregnant times, post-menopause, pre-menopause, and strong positive expression of wild p53 and Ki67 (Antigen identified by monoclonal antibody Ki-67), as well as without mutant p53 expression. The rs10773771 CT/TT alleles were identified to have a protective effect on EOC in women aged 53 years or older, as well as in cases with metastasis, advanced clinical stage, high pathological grade, multiple tumors, tumor size equal to or less than 3 cm, history of pregnancy, post-menopausal status, and strong positive expression of ER, PR, wild-type p53, PAX8, WT1, P16, and Ki67. Furthermore, rs10773771 CT/TT also showed a protective effect in patients with negative or mildly positive expression of PR, PAX8, wild-type p53, WT1, and P16, as well as positive expression of mutant p53. Compared to the reference haplotype GCG, individuals harboring haplotypes GTG were found to have a significantly decreased susceptibility to EOC. PIWIL1 was significantly expressed in the thyroid, pituitary, and adrenal glands with rs7957349 CC alleles. CONCLUSIONS PIWIL1 rs10848087 and rs7957349 were associated with increased risk of EOC, while rs10773771 may have a protective effect against EOC. These genetic variants may serve as potential biomarkers for EOC susceptibility in the South China population.
Collapse
Affiliation(s)
- Shanshan Liu
- Department of Hematology and Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China
| | - Yaping Yan
- Department of Hematology and Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China
| | - Zhizhong Cui
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong Province, China
| | - Haipeng Feng
- Department of Pathology, Shunde Hospital, Southern Medical University, Foshan, 528000, Guangdong, China
| | - Fengmei Zhong
- Department of Pathology, Shunde Hospital, Southern Medical University, Foshan, 528000, Guangdong, China
| | - Ziguang Liu
- Department of Pathology, Shunde Hospital, Southern Medical University, Foshan, 528000, Guangdong, China
| | - Yan Li
- Medical Research Center, Shunde Hospital, Southern Medical University, Foshan, 528000, Guangdong, China
| | - Xiang Ou
- The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China.
| | - Wenjuan Li
- Medical Research Center, Shunde Hospital, Southern Medical University, Foshan, 528000, Guangdong, China.
| |
Collapse
|
18
|
Wei X, Oxley S, Sideris M, Kalra A, Brentnall A, Sun L, Yang L, Legood R, Manchanda R. Quality of life after risk-reducing surgery for breast and ovarian cancer prevention: a systematic review and meta-analysis. Am J Obstet Gynecol 2023; 229:388-409.e4. [PMID: 37059410 DOI: 10.1016/j.ajog.2023.03.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/16/2023]
Abstract
OBJECTIVE This study aimed to assess the impact of risk-reducing surgery for breast cancer and ovarian cancer prevention on quality of life. We considered risk-reducing mastectomy, risk-reducing salpingo-oophorectomy, and risk-reducing early salpingectomy and delayed oophorectomy. DATA SOURCES We followed a prospective protocol (International Prospective Register of Systematic Reviews: CRD42022319782) and searched MEDLINE, Embase, PubMed, and Cochrane Library from inception to February 2023. STUDY ELIGIBILITY CRITERIA We followed a PICOS (population, intervention, comparison, outcome, and study design) framework. The population included women at increased risk of breast cancer or ovarian cancer. We focused on studies reporting quality of life outcomes (health-related quality of life, sexual function, menopause symptoms, body image, cancer-related distress or worry, anxiety, or depression) after risk-reducing surgery, including risk-reducing mastectomy for breast cancer and risk-reducing salpingo-oophorectomy or risk-reducing early salpingectomy and delayed oophorectomy for ovarian cancer. METHODS We used the Methodological Index for Non-Randomized Studies (MINORS) for study appraisal. Qualitative synthesis and fixed-effects meta-analysis were performed. RESULTS A total of 34 studies were included (risk-reducing mastectomy: 16 studies; risk-reducing salpingo-oophorectomy: 19 studies; risk-reducing early salpingectomy and delayed oophorectomy: 2 studies). Health-related quality of life was unchanged or improved in 13 of 15 studies after risk-reducing mastectomy (N=986) and 10 of 16 studies after risk-reducing salpingo-oophorectomy (N=1617), despite short-term deficits (N=96 after risk-reducing mastectomy and N=459 after risk-reducing salpingo-oophorectomy). Sexual function (using the Sexual Activity Questionnaire) was affected in 13 of 16 studies (N=1400) after risk-reducing salpingo-oophorectomy in terms of decreased sexual pleasure (-1.21 [-1.53 to -0.89]; N=3070) and increased sexual discomfort (1.12 [0.93-1.31]; N=1400). Hormone replacement therapy after premenopausal risk-reducing salpingo-oophorectomy was associated with an increase (1.16 [0.17-2.15]; N=291) in sexual pleasure and a decrease (-1.20 [-1.75 to -0.65]; N=157) in sexual discomfort. Sexual function was affected in 4 of 13 studies (N=147) after risk-reducing mastectomy, but stable in 9 of 13 studies (N=799). Body image was unaffected in 7 of 13 studies (N=605) after risk-reducing mastectomy, whereas 6 of 13 studies (N=391) reported worsening. Increased menopause symptoms were reported in 12 of 13 studies (N=1759) after risk-reducing salpingo-oophorectomy with a reduction (-1.96 [-2.81 to -1.10]; N=1745) in the Functional Assessment of Cancer Therapy - Endocrine Symptoms. Cancer-related distress was unchanged or decreased in 5 of 5 studies after risk-reducing mastectomy (N=365) and 8 of 10 studies after risk-reducing salpingo-oophorectomy (N=1223). Risk-reducing early salpingectomy and delayed oophorectomy (2 studies, N=413) led to better sexual function and menopause-specific quality of life. CONCLUSION Risk-reducing surgery may be associated with quality of life outcomes. Risk-reducing mastectomy and risk-reducing salpingo-oophorectomy reduce cancer-related distress, and do not affect health-related quality of life. Women and clinicians should be aware of body image problems after risk-reducing mastectomy, and of sexual dysfunction and menopause symptoms after risk-reducing salpingo-oophorectomy. Risk-reducing early salpingectomy and delayed oophorectomy may be a promising alternative to mitigate quality of life-related risks of risk-reducing salpingo-oophorectomy.
Collapse
Affiliation(s)
- Xia Wei
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London, United Kingdom; Wolfson Institute of Population Health, Cancer Research UK Barts Centre, Queen Mary University of London, London, United Kingdom
| | - Samuel Oxley
- Wolfson Institute of Population Health, Cancer Research UK Barts Centre, Queen Mary University of London, London, United Kingdom; Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London, United Kingdom
| | - Michail Sideris
- Wolfson Institute of Population Health, Cancer Research UK Barts Centre, Queen Mary University of London, London, United Kingdom; Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London, United Kingdom
| | - Ashwin Kalra
- Wolfson Institute of Population Health, Cancer Research UK Barts Centre, Queen Mary University of London, London, United Kingdom; Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London, United Kingdom
| | - Adam Brentnall
- Wolfson Institute of Population Health, Cancer Research UK Barts Centre, Queen Mary University of London, London, United Kingdom
| | - Li Sun
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London, United Kingdom; Wolfson Institute of Population Health, Cancer Research UK Barts Centre, Queen Mary University of London, London, United Kingdom
| | - Li Yang
- School of Public Health, Peking University, Beijing, China
| | - Rosa Legood
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Ranjit Manchanda
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London, United Kingdom; Wolfson Institute of Population Health, Cancer Research UK Barts Centre, Queen Mary University of London, London, United Kingdom; Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London, United Kingdom; Medical Research Council Clinical Trials Unit, Institute of Clinical Trials and Methodology, Faculty of Population Health Sciences, University College London, London, United Kingdom; Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
19
|
Wang T, Dossett LA. Incorporating Value-Based Decisions in Breast Cancer Treatment Algorithms. Surg Oncol Clin N Am 2023; 32:777-797. [PMID: 37714643 DOI: 10.1016/j.soc.2023.05.008] [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: 09/17/2023]
Abstract
Given the excellent prognosis and availability of evidence-based treatment, patients with early-stage breast cancer are at risk of overtreatment. In this review, we summarize key opportunities to incorporate value-based decisions to optimize the delivery of high-value treatment across the breast cancer care continuum.
Collapse
Affiliation(s)
- Ton Wang
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lesly A Dossett
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Institute for Healthcare Policy and Innovation, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
20
|
Abdel-Razeq H, Abbasi S, Abdeen G, Abdulelah H, Debs J, Al Masri S, Aljadayeh MH, Awidi A. Management of breast cancer patients with BRCA gene mutations in Jordan: perspectives and challenges. Hosp Pract (1995) 2023; 51:184-191. [PMID: 37927299 DOI: 10.1080/21548331.2023.2266019] [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: 01/29/2023] [Accepted: 09/28/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND This paper explores and discusses local challenges oncologists face for diagnosing and managing breast cancer patients with BRCA gene mutations in Jordan. METHODS A task force involving key opinion leaders, experts in the management of breast cancer, and stakeholders in healthcare systems where genetic testing is available in Jordan discussed current evidence and local real-life practice. The task force then formulated recommendations to achieve better patient outcomes and satisfaction based on evidence-based medicine and their clinical experience in BRCA-mutated breast cancer management. RESULTS AND CONCLUSION Eligibility of patients for genetic testing, physician acceptance and willingness to integrate genetic testing into routine practice is encouraging but remains restricted by testing availability and financial coverage. Until more data is available, genetic testing should be targeted for breast cancer patients based on tumor subtypes, as well as family and personal history of cancer, as per international guidelines. Whenever possible, genetic testing should aim to detect all actionable genes through a multigene panel including BRCA1/2. Major challenges faced in clinical practice in Jordan include fear of genetic discrimination and social stigmatization, as well as hesitancy toward risk-reducing surgery. Pre-testing counseling is therefore critical to promote acceptance of genetic testing. Since geneticists are in short supply in Jordan, genetic counseling can be offered through a specially trained genetic counselor or through a hybrid system that includes oncologist-based counselling. In addition to cancer prevention, germline genetic testing may assist in the selection of specific anti-cancer therapy, such as PARP inhibitors, in patients with BRCA1/2 mutation. Nationwide initiatives are also needed to ensure access to PARP inhibition therapy and provide financial coverage for genetic screening, mastectomies and reconstructive surgery across Jordan.
Collapse
Affiliation(s)
- Hikmat Abdel-Razeq
- Deputy Director General, Chief Medical Officer, King Hussein Cancer Center, Amman, Jordan
| | - Salah Abbasi
- Hematology and Medical Oncology; Associate Professor of Medicine, Hematology & Oncology, Private Sector, Jordan University, Amman, Jordan
| | - Ghadeer Abdeen
- Consultant Internal Medicine and Medical Oncologist, King Hussein Cancer Center, Amman, Jordan
| | - Hazem Abdulelah
- Consultant Internal Medicine and Medical Oncologist, King Hussein Cancer Center, Amman, Jordan
| | - Jamil Debs
- Medical Affairs, Pfizer Inc., Beirut, Lebanon
| | | | - Majdi H Aljadayeh
- Head of Medical Oncology, Hematology and Stem Cell Transplant Unit, Military Cancer Center, Royal Medical Services, Amman, Jordan
| | - Abdalla Awidi
- Professor of Medicine, Hematology & Oncology, Director of cell therapy center, Chief of Hematology & Oncology, Cell Therapy center, The University of Jordan, Amman, Jordan
| |
Collapse
|
21
|
Fahim SM, Alexander CSW, Qian J, Ngorsuraches S, Hohmann NS, Lloyd KB, Reagan A, Hart L, McCormick N, Westrick SC. Current published evidence on barriers and proposed strategies for genetic testing implementation in health care settings: A scoping review. J Am Pharm Assoc (2003) 2023; 63:998-1016. [PMID: 37119989 DOI: 10.1016/j.japh.2023.04.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 04/20/2023] [Accepted: 04/22/2023] [Indexed: 05/01/2023]
Abstract
BACKGROUND The slow uptake of genetic testing in routine clinical practice warrants the attention of researchers and practitioners to find effective strategies to facilitate implementation. OBJECTIVES This study aimed to identify the barriers to and strategies for pharmacogenetic testing implementation in a health care setting from published literature. METHODS A scoping review was conducted in August 2021 with an expanded literature search using Ovid MEDLINE, Web of Science, International Pharmaceutical Abstract, and Google Scholar to identify studies reporting implementation of pharmacogenetic testing in a health care setting, from a health care system's perspective. Articles were screened using DistillerSR and findings were organized using the 5 major domains of Consolidated Framework for Implementation Research (CFIR). RESULTS A total of 3536 unique articles were retrieved from the above sources, with only 253 articles retained after title and abstract screening. Upon screening the full texts, 57 articles (representing 46 unique practice sites) were found matching the inclusion criteria. We found that most reported barriers and their associated strategies to the implementation of pharmacogenetic testing surrounded 2 CFIR domains: intervention characteristics and inner settings. Factors relating to cost and reimbursement were described as major barriers in the intervention characteristics. In the same domain, another major barrier was the lack of utility studies to provide evidence for genetic testing uptake. Technical hurdles, such as integrating genetic information to medical records, were identified as an inner settings barrier. Collaborations and lessons from early implementers could be useful strategies to overcome majority of the barriers across different health care settings. Strategies proposed by the included implementation studies to overcome these barriers are summarized and can be used as guidance in future. CONCLUSION Barriers and strategies identified in this scoping review can provide implementation guidance for practice sites that are interested in implementing genetic testing.
Collapse
|
22
|
Tu SH, Huang WT, Chew CH, Chen AL, Chen ST, Chen JH, Hsieh YC, Chen CC. Unveiling the Power of Anticancer Drug Screening: A Clinical Case Study Comparing the Effectiveness of Hollow Fiber Assay Microtube Array Membrane (MTAM-HFA) in Breast Cancer Patients. Cancers (Basel) 2023; 15:2764. [PMID: 37345100 DOI: 10.3390/cancers15102764] [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: 04/12/2023] [Revised: 05/02/2023] [Accepted: 05/06/2023] [Indexed: 06/23/2023] Open
Abstract
Breast cancer is a severe public health problem, and early treatment with powerful anticancer drugs is critical for success. The researchers investigated the clinical results of a novel screening tool termed Microtube Array Membrane Hollow Fiber Assay (MTAM-HFA) in breast cancer patients in this clinical investigation. In all trial participants, the MTAM-HFA was utilized to identify active medicines for the treatment of breast cancer. The MTAM-HFA was shown to be extremely useful in predicting patient response to anticancer medication therapy in this study. Furthermore, the substantial association between the MTAM-HFA screening outcome and the clinical outcome of the respective patients emphasizes the promise of this unique screening technology in discovering effective anticancer medication combinations for the treatment of breast cancer. These findings indicate that the MTAM-HFA has clinical significance and might be a valuable tool in the development of tailored therapy for cancer care. This study provides helpful information for physicians and scientists working on breast cancer therapy research. The potential benefits of employing MTAM-HFA to find accurate therapies for breast cancer patients might lead to enhanced personalized medicine approaches to cancer care, resulting in better patient outcomes. Overall, the MTAM-HFA screening approach has the potential to revolutionize customized cancer therapy, providing hope to both patients and physicians.
Collapse
Affiliation(s)
- Shih-Hsin Tu
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Surgery, Taipei Medical University Hospital, Taipei 11052, Taiwan
| | - Wan-Ting Huang
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei 11052, Taiwan
| | - Chee Ho Chew
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei 11052, Taiwan
| | - Amanda Lin Chen
- Translational Autoinflammatory Disease Section (TADS), Laboratory of Clinical Immunology and Microbiology (LCIM), National Institutes of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Shou-Tung Chen
- Comprehensive Breast Cancer Center, Changhua Christian Hospital, Changhua 50094, Taiwan
- Department of Medical Research, Changhua Christian Hospital, Changhua 50094, Taiwan
| | - Jin-Hua Chen
- Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei 11052, Taiwan
| | - Yi-Chen Hsieh
- Ph.D. Program in Medical Neuroscience, Taipei Medical University, Taipei 250, Taiwan
| | - Chien-Chung Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, Taipei 11052, Taiwan
- Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei 250, Taiwan
| |
Collapse
|
23
|
Guzauskas GF, Garbett S, Zhou Z, Schildcrout JS, Graves JA, Williams MS, Hao J, Jones LK, Spencer SJ, Jiang S, Veenstra DL, Peterson JF. Population Genomic Screening for Three Common Hereditary Conditions : A Cost-Effectiveness Analysis. Ann Intern Med 2023; 176:585-595. [PMID: 37155986 DOI: 10.7326/m22-0846] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND The cost-effectiveness of screening the U.S. population for Centers for Disease Control and Prevention (CDC) Tier 1 genomic conditions is unknown. OBJECTIVE To estimate the cost-effectiveness of simultaneous genomic screening for Lynch syndrome (LS), hereditary breast and ovarian cancer syndrome (HBOC), and familial hypercholesterolemia (FH). DESIGN Decision analytic Markov model. DATA SOURCES Published literature. TARGET POPULATION Separate age-based cohorts (ages 20 to 60 years at time of screening) of racially and ethnically representative U.S. adults. TIME HORIZON Lifetime. PERSPECTIVE U.S. health care payer. INTERVENTION Population genomic screening using clinical sequencing with a restricted panel of high-evidence genes, cascade testing of first-degree relatives, and recommended preventive interventions for identified probands. OUTCOME MEASURES Incident breast, ovarian, and colorectal cancer cases; incident cardiovascular events; quality-adjusted survival; and costs. RESULTS OF BASE-CASE ANALYSIS Screening 100 000 unselected 30-year-olds resulted in 101 (95% uncertainty interval [UI], 77 to 127) fewer overall cancer cases and 15 (95% UI, 4 to 28) fewer cardiovascular events and an increase of 495 quality-adjusted life-years (QALYs) (95% UI, 401 to 757) at an incremental cost of $33.9 million (95% UI, $27.0 million to $41.1 million). The incremental cost-effectiveness ratio was $68 600 per QALY gained (95% UI, $41 800 to $88 900). RESULTS OF SENSITIVITY ANALYSIS Screening 30-, 40-, and 50-year-old cohorts was cost-effective in 99%, 88%, and 19% of probabilistic simulations, respectively, at a $100 000-per-QALY threshold. The test costs at which screening 30-, 40-, and 50-year-olds reached the $100 000-per-QALY threshold were $413, $290, and $166, respectively. Variant prevalence and adherence to preventive interventions were also highly influential parameters. LIMITATIONS Population averages for model inputs, which were derived predominantly from European populations, vary across ancestries and health care environments. CONCLUSION Population genomic screening with a restricted panel of high-evidence genes associated with 3 CDC Tier 1 conditions is likely to be cost-effective in U.S. adults younger than 40 years if the testing cost is relatively low and probands have access to preventive interventions. PRIMARY FUNDING SOURCE National Human Genome Research Institute.
Collapse
Affiliation(s)
- Gregory F Guzauskas
- The CHOICE Institute, Department of Pharmacy, University of Washington, Seattle, Washington (G.F.G., S.J.)
| | - Shawn Garbett
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee (S.G., J.S.S.)
| | - Zilu Zhou
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, Tennessee (Z.Z., J.A.G.)
| | - Jonathan S Schildcrout
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee (S.G., J.S.S.)
| | - John A Graves
- Department of Health Policy, Vanderbilt University Medical Center, Nashville, Tennessee (Z.Z., J.A.G.)
| | - Marc S Williams
- Department of Genomic Health, Geisinger, Danville, Pennsylvania (M.S.W.)
| | - Jing Hao
- Department of Genomic Health and Department of Population Health Sciences, Geisinger, Danville, Pennsylvania (J.H.)
| | - Laney K Jones
- Department of Population Health Sciences and Heart Institute, Geisinger, Danville, Pennsylvania (L.K.J.)
| | - Scott J Spencer
- Institute for Public Health Genetics, University of Washington, Seattle, Washington (S.J.S.)
| | - Shangqing Jiang
- The CHOICE Institute, Department of Pharmacy, University of Washington, Seattle, Washington (G.F.G., S.J.)
| | - David L Veenstra
- The CHOICE Institute, Department of Pharmacy, and Institute for Public Health Genetics, University of Washington, Seattle, Washington (D.L.V.)
| | - Josh F Peterson
- Department of Biomedical Informatics and Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee (J.F.P.)
| |
Collapse
|
24
|
De Silva DL, Stafford L, Skandarajah AR, Sinclair M, Devereux L, Hogg K, Kentwell M, Park A, Lal L, Zethoven M, Jayawardana MW, Chan F, Butow PN, James PA, Mann GB, Campbell IG, Lindeman GJ. Universal genetic testing for women with newly diagnosed breast cancer in the context of multidisciplinary team care. Med J Aust 2023; 218:368-373. [PMID: 37005005 PMCID: PMC10952347 DOI: 10.5694/mja2.51906] [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: 09/07/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 04/04/2023]
Abstract
OBJECTIVE To determine the feasibility of universal genetic testing of women with newly diagnosed breast cancer, to estimate the incidence of pathogenic gene variants and their impact on patient management, and to evaluate patient and clinician acceptance of universal testing. DESIGN, SETTING, PARTICIPANTS Prospective study of women with invasive or high grade in situ breast cancer and unknown germline status discussed at the Parkville Breast Service (Melbourne) multidisciplinary team meeting. Women were recruited to the pilot (12 June 2020 - 22 March 2021) and expansion phases (17 October 2021 - 8 November 2022) of the Mutational Assessment of newly diagnosed breast cancer using Germline and tumour genomICs (MAGIC) study. MAIN OUTCOME MEASURES Germline testing by DNA sequencing, filtered for nineteen hereditary breast and ovarian cancer genes that could be classified as actionable; only pathogenic variants were reported. Surveys before and after genetic testing assessed pilot phase participants' perceptions of genetic testing, and psychological distress and cancer-specific worry. A separate survey assessed clinicians' views on universal testing. RESULTS Pathogenic germline variants were identified in 31 of 474 expanded study phase participants (6.5%), including 28 of 429 women with invasive breast cancer (6.5%). Eighteen of the 31 did not meet current genetic testing eligibility guidelines (probability of a germline pathogenic variant ≥ 10%, based on CanRisk, or Manchester score ≥ 15). Clinical management was changed for 24 of 31 women after identification of a pathogenic variant. Including 68 further women who underwent genetic testing outside the study, 44 of 542 women carried pathogenic variants (8.1%). Acceptance of universal testing was high among both patients (90 of 103, 87%) and clinicians; no decision regret or adverse impact on psychological distress or cancer-specific worry were reported. CONCLUSION Universal genetic testing following the diagnosis of breast cancer detects clinically significant germline pathogenic variants that might otherwise be missed because of testing guidelines. Routine testing and reporting of pathogenic variants is feasible and acceptable for both patients and clinicians.
Collapse
Affiliation(s)
- Dilanka L De Silva
- The University of MelbourneMelbourneVIC
- Parkville Familial Cancer CentrePeter MacCallum Cancer Centre and Royal Melbourne HospitalMelbourneVIC
- Memorial Sloan Kettering Cancer CenterNew YorkNYUnited States of America
| | - Lesley Stafford
- The University of MelbourneMelbourneVIC
- The Royal Melbourne HospitalMelbourneVIC
| | - Anita R Skandarajah
- The University of MelbourneMelbourneVIC
- The Royal Melbourne HospitalMelbourneVIC
| | | | - Lisa Devereux
- The University of MelbourneMelbourneVIC
- Peter MacCallum Cancer CentreMelbourneVIC
| | - Kirsten Hogg
- The University of MelbourneMelbourneVIC
- Walter and Eliza Hall Institute of Medical ResearchMelbourneVIC
| | - Maira Kentwell
- The University of MelbourneMelbourneVIC
- Parkville Familial Cancer CentrePeter MacCallum Cancer Centre and Royal Melbourne HospitalMelbourneVIC
| | - Allan Park
- The Royal Melbourne HospitalMelbourneVIC
| | - Luxi Lal
- The Royal Melbourne HospitalMelbourneVIC
- Peter MacCallum Cancer CentreMelbourneVIC
- Walter and Eliza Hall Institute of Medical ResearchMelbourneVIC
| | | | - Madawa W Jayawardana
- The University of MelbourneMelbourneVIC
- Peter MacCallum Cancer CentreMelbourneVIC
| | - Fiona Chan
- The Royal Children's Hospital MelbourneMelbourneVIC
| | - Phyllis N Butow
- Centre for Medical Psychology and Evidence‐based Decision Making, the University of SydneySydneyNSW
| | - Paul A James
- The University of MelbourneMelbourneVIC
- Parkville Familial Cancer CentrePeter MacCallum Cancer Centre and Royal Melbourne HospitalMelbourneVIC
- The Royal Melbourne HospitalMelbourneVIC
- Peter MacCallum Cancer CentreMelbourneVIC
| | - G Bruce Mann
- The University of MelbourneMelbourneVIC
- The Royal Melbourne HospitalMelbourneVIC
- Royal Women's HospitalMelbourneVIC
| | - Ian G Campbell
- The University of MelbourneMelbourneVIC
- Peter MacCallum Cancer CentreMelbourneVIC
| | - Geoffrey J Lindeman
- The University of MelbourneMelbourneVIC
- Parkville Familial Cancer CentrePeter MacCallum Cancer Centre and Royal Melbourne HospitalMelbourneVIC
- Peter MacCallum Cancer CentreMelbourneVIC
- Walter and Eliza Hall Institute of Medical ResearchMelbourneVIC
| |
Collapse
|
25
|
Huang Y, Huang X, Huang X, Lin S, Luo S, Gu D, Weng X, Xu X. Cost-effectiveness analysis of ovarian function preservation with GnRH agonist during chemotherapy in premenopausal women with early breast cancer. Hum Reprod 2023:7131352. [PMID: 37075316 DOI: 10.1093/humrep/dead073] [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: 06/23/2022] [Revised: 03/25/2023] [Indexed: 04/21/2023] Open
Abstract
STUDY QUESTION Is it economically worthwhile to use GnRH agonist (GnRHa) to prevent menopausal symptoms (MS) and protect fertility in premenopausal women with breast cancer (BC) during chemotherapy from the US perspective? SUMMARY ANSWER It is cost-effective to administer GnRHa during chemotherapy in order to forefend MS in premenopausal patients with BC when the willingness-to-pay (WTP) threshold is $50 000.00 per quality-adjusted life-year (QALY), and to preserve fertility in young patients with BC who undergo oocyte cryopreservation (OC), or no OC, when the WTP thresholds per live birth are $71 333.33 and $61 920.00, respectively. WHAT IS KNOWN ALREADY Chemotherapy often results in premature ovarian insufficiency (POI) in premenopausal survivors of BC, causing MS and infertility. Administering GnRHa during chemotherapy has been recommended for ovarian function preservation by international guidelines. STUDY DESIGN, SIZE, DURATION Two decision-analytic models were developed, respectively, for preventing MS and protecting fertility over a 5-year period, which compared the cost-effectiveness of two strategies: adding GnRHa during chemotherapy (GnRHa plus Chemo) or chemotherapy alone (Chemo). PARTICIPANTS/MATERIALS, SETTING, METHODS The participants were early premenopausal women with BC aged 18-49 years who were undergoing chemotherapy. Two decision tree models were constructed: one for MS prevention and one for fertility protection from the US perspective. All data were obtained from published literature and official websites. The models' primary outcomes included QALYs and incremental cost-effectiveness ratios (ICERs). The robustness of the models was tested by sensitivity analyses. MAIN RESULTS AND THE ROLE OF CHANCE In the MS model, GnRHa plus Chemo resulted in an ICER of $17 900.85 per QALY compared with Chemo, which was greater than the WTP threshold of $50 000.00 per QALY; therefore, GnRHa plus Chemo was a cost-effective strategy for premenopausal women with BC in the USA. Probabilistic sensitivity analysis (PSA) results showed an 81.76% probability of cost-effectiveness in the strategy. In the fertility model, adding GnRHa for patients undergoing OC and those who were unable to undergo OC resulted in ICERs of $67 933.50 and $60 209.00 per live birth in the USA, respectively. PSA indicated that GnRHa plus Chemo was more likely to be cost-effective over Chemo when the WTP for an additional live birth exceed $71 333.33 in Context I (adding GnRHa to preserve fertility in young patients with BC after OC) and $61 920.00 in Context II (adding GnRHa to preserve fertility in young patients with BC who cannot accept OC). LIMITATIONS, REASONS FOR CAUTION The indirect costs, such as disease-related mental impairment and non-medical costs (e.g. transportation cost) were not included. All data were derived from previously published literature and databases, which might yield some differences from the real world. In addition, the POI-induced MS with a lower prevalence and the specific strategy of chemotherapy were not considered in the MS model, and the 5-year time horizon for having a child might not be suitable for all patients in the fertility model. WIDER IMPLICATIONS OF THE FINDINGS When considering the economic burden of cancer survivors, the results of this study provide an evidence-based reference for clinical decision-making, showing that it is worthwhile to employ GnRHa during chemotherapy to prevent MS and preserve fertility. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the Natural Science Foundation of Fujian Province [2021J02038]; and the Startup Fund for Scientific Research, Fujian Medical University [2021QH1059]. All authors declare no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Yaping Huang
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaoting Huang
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaojia Huang
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shen Lin
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shaohong Luo
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Dian Gu
- Institute for Health & Aging, University of California, San Francisco, CA, USA
| | - Xiuhua Weng
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiongwei Xu
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
26
|
Jiang J, Jiang S, Ahumada-Canale A, Chen Z, Si L, Jiang Y, Yang L, Gu Y. Breast Cancer Screening Should Embrace Precision Medicine: Evidence by Reviewing Economic Evaluations in China. Adv Ther 2023; 40:1393-1417. [PMID: 36800077 PMCID: PMC10070309 DOI: 10.1007/s12325-023-02450-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/01/2023] [Indexed: 02/18/2023]
Abstract
The cost-effectiveness of conventional population-based breast cancer screening strategies (e.g. mammography) has been found controversial, while evidence shows that genetic testing for early detection of pathogenic variants is cost-effective. We aimed to review the economic evaluations of breast cancer screening in China to provide an information summary for future research on this topic. We searched the literature to identify the economic evaluations that examined breast cancer screening and testing in China, supplemented by hand-searching the reference lists of the included studies. We finally included five studies satisfying our inclusion criteria. Four articles examined mammography while the rest investigated multigene testing. The existing breast cancer screening programmes were found to be cost-effective among urban Chinese women, but one study concluded that they might cause harm to women in rural areas. Contextual factors, such as data absence, urban-rural disparity, willingness-to-pay threshold, and model design, imposed barriers to cost-effectiveness analysis. Multigene testing was found to be cost-effective and has a promising population impact among all women with breast cancer in China. Future research should investigate the cost-effectiveness of screening and identifying breast cancer through precision medicine technologies, including genetic testing, genome sequencing, cascade testing, and the return of secondary findings.
Collapse
Affiliation(s)
- Jingjing Jiang
- Health Economics Research Centre, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Shan Jiang
- School of Population and Public Health, The University of British Columbia, Vancouver, BC, Canada
| | - Antonio Ahumada-Canale
- Macquarie University Centre for the Health Economy, Macquarie Business School & Australian Institute of Health Innovation, Macquarie University, Sydney, NSW, Australia
| | - Zhuo Chen
- Department of Health Policy and Management, College of Public Health, University of Georgia, Athens, GA, USA
- Faculty of Humanities and Social Sciences, School of Economics, University of Nottingham Ningbo China, Ningbo, Zhejiang, China
| | - Lei Si
- School of Health Sciences, Western Sydney University, Campbelltown, Australia
- Translational Health Research Institute, Western Sydney University, Penrith, Australia
| | - Yawen Jiang
- School of Public Health (Shenzhen), Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Li Yang
- School of Public Health, Peking University, Beijing, China.
| | - Yuanyuan Gu
- Macquarie University Centre for the Health Economy, Macquarie Business School & Australian Institute of Health Innovation, Macquarie University, Sydney, NSW, Australia.
| |
Collapse
|
27
|
Lang N, Ayme A, Ming C, Combes JD, Chappuis VN, Friedlaender A, Vuilleumier A, Sandoval JL, Viassolo V, Chappuis PO, Labidi-Galy SI. Chemotherapy-related agranulocytosis as a predictive factor for germline BRCA1 pathogenic variants in breast cancer patients: a retrospective cohort study. Swiss Med Wkly 2023; 153:40055. [PMID: 37011610 DOI: 10.57187/smw.2023.40055] [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] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND Carriers of germline pathogenic variants of the BRCA1 gene (gBRCA1) tend to have a higher incidence of haematological toxicity upon exposure to chemotherapy. We hypothesised that the occurrence of agranulocytosis during the first cycle of (neo-)adjuvant chemotherapy (C1) in breast cancer (BC) patients could predict gBRCA1 pathogenic variants. PATIENTS AND METHODS The study population included non-metastatic BC patients selected for genetic counselling at Hôpitaux Universitaires de Genève (Jan. 1998 to Dec. 2017) with available mid-cycle blood counts performed during C1. The BOADICEA and Manchester scoring system risk-prediction models were applied. The primary outcome was the predicted likelihood of harbouring gBRCA1 pathogenic variants among patients presenting agranulocytosis during C1. RESULTS Three hundred seven BC patients were included: 32 (10.4%) gBRCA1, 27 (8.8%) gBRCA2, and 248 (81.1%) non-heterozygotes. Mean age at diagnosis was 40 years. Compared with non-heterozygotes, gBRCA1 heterozygotes more frequently had grade 3 BC (78.1%; p = 0.014), triple-negative subtype (68.8%; p <0.001), bilateral BC (25%; p = 0.004), and agranulocytosis following the first cycle of (neo-)adjuvant chemotherapy (45.8%; p = 0.002). Agranulocytosis and febrile neutropenia that developed following the first cycle of chemotherapy were independently predictive for gBRCA1 pathogenic variants (odds ratio: 6.1; p = 0.002). The sensitivity, specificity, positive predictive value, and negative predictive value for agranulocytosis predicting gBRCA1 were 45.8% (25.6-67.2%), 82.8% (77.5-87.3%), 22.9% (6.1-37.3%), and 93.4% (88.9-96.4%), respectively. Agranulocytosis substantially improved the positive predictive value of the risk-prediction models used for gBRCA1 evaluation. CONCLUSION Agranulocytosis following the first cycle of (neo-)adjuvant chemotherapy is an independent predictive factor for gBRCA1 detection in non-metastatic BC patients.
Collapse
Affiliation(s)
- Noémie Lang
- Department of Oncology, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Aurélie Ayme
- Department of Diagnostics, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Chang Ming
- Department of Clinical Research, Faculty of Medicine, University of Basel, Basel, Switzerland
| | - Jean-Damien Combes
- Infections and Cancer Epidemiology Group, International Agency for Research on Cancer, Lyon, France
| | - Victor N Chappuis
- Department of Oncology, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Alex Friedlaender
- Department of Oncology, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Aurélie Vuilleumier
- Department of Oncology, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - José L Sandoval
- Department of Oncology, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Valeria Viassolo
- Department of Oncology, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Pierre O Chappuis
- Department of Oncology, Hôpitaux Universitaires de Genève, Geneva, Switzerland
- Department of Diagnostics, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - S Intidhar Labidi-Galy
- Department of Oncology, Hôpitaux Universitaires de Genève, Geneva, Switzerland
- Department of Diagnostics, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| |
Collapse
|
28
|
Stoltze UK, Hagen CM, van Overeem Hansen T, Byrjalsen A, Gerdes AM, Yakimov V, Rasmussen S, Bækvad-Hansen M, Hougaard DM, Schmiegelow K, Hjalgrim H, Wadt K, Bybjerg-Grauholm J. Combinatorial batching of DNA for ultralow-cost detection of pathogenic variants. Genome Med 2023; 15:17. [PMID: 36918911 PMCID: PMC10013285 DOI: 10.1186/s13073-023-01167-6] [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: 11/12/2022] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND Next-generation sequencing (NGS) based population screening holds great promise for disease prevention and earlier diagnosis, but the costs associated with screening millions of humans remain prohibitive. New methods for population genetic testing that lower the costs of NGS without compromising diagnostic power are needed. METHODS We developed double batched sequencing where DNA samples are batch-sequenced twice - directly pinpointing individuals with rare variants. We sequenced batches of at-birth blood spot DNA using a commercial 113-gene panel in an explorative (n = 100) and a validation (n = 100) cohort of children who went on to develop pediatric cancers. All results were benchmarked against individual whole genome sequencing data. RESULTS We demonstrated fully replicable detection of cancer-causing germline variants, with positive and negative predictive values of 100% (95% CI, 0.91-1.00 and 95% CI, 0.98-1.00, respectively). Pathogenic and clinically actionable variants were detected in RB1, TP53, BRCA2, APC, and 19 other genes. Analyses of larger batches indicated that our approach is highly scalable, yielding more than 95% cost reduction or less than 3 cents per gene screened for rare disease-causing mutations. We also show that double batched sequencing could cost-effectively prevent childhood cancer deaths through broad genomic testing. CONCLUSIONS Our ultracheap genetic diagnostic method, which uses existing sequencing hardware and standard newborn blood spots, should readily open up opportunities for population-wide risk stratification using genetic screening across many fields of clinical genetics and genomics.
Collapse
Affiliation(s)
- Ulrik Kristoffer Stoltze
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Blegdamsvej 9, 2100, KBH Ø, Denmark. .,Department of Clinical Genetics, Rigshospitalet, Blegdamsvej 9, 2100, KBH Ø, Denmark.
| | - Christian Munch Hagen
- Department of Congenital Disorders, Statens Serum Institute, 2300, KBH S, Artillerivej 5, Denmark
| | - Thomas van Overeem Hansen
- Department of Clinical Genetics, Rigshospitalet, Blegdamsvej 9, 2100, KBH Ø, Denmark.,Department of Clinical Medicine, Copenhagen University, Blegdamsvej 3B, 2200, KBH N, Denmark
| | - Anna Byrjalsen
- Department of Clinical Genetics, Rigshospitalet, Blegdamsvej 9, 2100, KBH Ø, Denmark
| | - Anne-Marie Gerdes
- Department of Clinical Genetics, Rigshospitalet, Blegdamsvej 9, 2100, KBH Ø, Denmark
| | - Victor Yakimov
- Department of Congenital Disorders, Statens Serum Institute, 2300, KBH S, Artillerivej 5, Denmark
| | - Simon Rasmussen
- Novo Nordisk Foundation Center for Protein Research, Copenhagen University, Blegdamsvej 3B, 2200, KBH N, Denmark
| | - Marie Bækvad-Hansen
- Department of Congenital Disorders, Statens Serum Institute, 2300, KBH S, Artillerivej 5, Denmark
| | - David Michael Hougaard
- Department of Congenital Disorders, Statens Serum Institute, 2300, KBH S, Artillerivej 5, Denmark
| | - Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Blegdamsvej 9, 2100, KBH Ø, Denmark.,Department of Clinical Medicine, Copenhagen University, Blegdamsvej 3B, 2200, KBH N, Denmark
| | - Henrik Hjalgrim
- Department of Clinical Medicine, Copenhagen University, Blegdamsvej 3B, 2200, KBH N, Denmark.,Danish Cancer Society Research Centre, Danish Cancer Society, Strandboulevarden 49, 2100, KBH Ø, Denmark.,Department of Epidemiology Research, Statens Serum Institut, 2300, KBH S, Artillerivej 5, Denmark.,Department of Haematology, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen Ø, Denmark
| | - Karin Wadt
- Department of Clinical Genetics, Rigshospitalet, Blegdamsvej 9, 2100, KBH Ø, Denmark
| | - Jonas Bybjerg-Grauholm
- Department of Congenital Disorders, Statens Serum Institute, 2300, KBH S, Artillerivej 5, Denmark.
| |
Collapse
|
29
|
Rezapour A, Souresrafil A, Barzegar M, Sheikhy-Chaman M, Tatarpour P. Economic evaluation of next-generation sequencing techniques in diagnosis of genetic disorders: A systematic review. Clin Genet 2023; 103:513-528. [PMID: 36808726 DOI: 10.1111/cge.14313] [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: 09/28/2022] [Revised: 02/12/2023] [Accepted: 02/14/2023] [Indexed: 02/23/2023]
Abstract
In recent years, massively parallel sequencing or next generation sequencing (NGS) has considerably changed both the research and diagnostic fields, and rapid developments have led to the combination of NGS techniques in clinical practice, ease of analysis, and detection of genetic mutations. This article aimed at reviewing the economic evaluation studies of the NGS techniques in the diagnosis of genetic diseases. In this systematic review, scientific databases (PubMed, EMBASE, Web of Science, Cochrane, Scopus, and CEA registry) were searched from 2005 to 2022 to identify the related literature on the economic evaluation of NGS techniques in the diagnosis of genetic diseases. Full-text reviews and data extraction were all performed by two independent researchers. The quality of all the articles included in this study was evaluated using the Checklist of Quality of Health Economic Studies (QHES). Out of 20 521 screened abstracts, 36 studies met the inclusion criteria. The mean score of the QHES checklist for the studies was 0.78 (high quality). Seventeen studies were conducted based on modeling. Cost-effectiveness analysis, cost-utility analysis, and cost-minimization analysis were done in 26 studies, 13 studies, and 1 study, respectively. Based on the available evidence and findings, exome sequencing, which is one of the NGS techniques, could have the potential to be used as a cost-effective genomic test to diagnose children with suspected genetic diseases. The results of the present study support the cost-effectiveness of exome sequencing in diagnosing suspected genetic disorders. However, the use of exome sequencing as a first- or second-line diagnostic test is still controversial. Most studies have been conducted in high-income countries, and research on the cost-effectiveness of NGS methods is recommended in low- and middle-income countries.
Collapse
Affiliation(s)
- Aziz Rezapour
- Health Management and Economics Research Center, Health Management Research Institute, Iran University of Medical Sciences, Tehran, Iran
| | - Aghdas Souresrafil
- Department of Health Services and Health Promotion, School of Health, Occupational Environment Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Barzegar
- Department of English Language, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Sheikhy-Chaman
- Department of Health Economics, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Parvin Tatarpour
- School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Robson M. Testing for Inherited Susceptibility to Breast Cancer. Hematol Oncol Clin North Am 2023; 37:17-31. [PMID: 36435609 DOI: 10.1016/j.hoc.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
When BRCA1 and BRCA2 were first identified, the initial models for delivering testing were shaped by concepts of genetic exceptionalism and a lack of data regarding therapeutic implications and the effectiveness of risk reduction. Since then, interventions have been effective, and treatment implications have become clear. The sensitivity of guideline-based testing is incomplete, leading to calls for universal testing. Completely universal testing, however, is not necessary to identify the great majority of BRCA1 or BRCA2 variants. Broader testing (both in terms of eligibility and genes tested) will identify more variants, particularly in moderate penetrance genes, but the clinical implications remain less clear for these variants.
Collapse
Affiliation(s)
- Mark Robson
- Breast Medicine Service, Department of Medicine, Memorial Hospital for Treatment of Cancer and Allied Disease, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, 300 East 66th Street, Room 813, New York, NY 10065, USA.
| |
Collapse
|
31
|
Ain Q, Richardson C, Mutebi M, George A, Kemp Z, Rusby JE. Does mainstream BRCA testing affect surgical decision-making in newly-diagnosed breast cancer patients? Breast 2023; 67:30-35. [PMID: 36577271 PMCID: PMC9982265 DOI: 10.1016/j.breast.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/22/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Germline pathogenic variants mutations) in the BRCA1 and BRCA2 genes cause an increased risk of breast cancer and ovarian cancer. Mainstream cancer genetic testing (MCG) was introduced for breast cancer patients in our unit in 2013. Non-geneticist clinicians have been trained to offer genetic testing during initial treatment planning. We assessed the impact of timely test results on surgical decision-making. METHODS Women who had undergone mainstream genetic testing for breast cancer between September 2013 and September 2018 were identified from a prospective database. Surgical data were collected retrospectively. RESULTS 580 eligible women had mainstream genetic testing. For 474 this was their first breast cancer diagnosis. The median age was 46 years (interquartile range (IQR) 38-57). The indications were: age ≤45 years for 233 (49%); triple negative disease for 192 women (40.5%); bilateral breast cancer age <60 for 39 (8%) and other for 72 (14%) women. The median time for test initiation to result was 18 days (IQR 15-21). 302 (64% received results before surgery. 88% of those found to have a BRCA mutation before surgery opted for bilateral mastectomy (compared to 5% with BRCA wild type). An additional 106 patients had a new diagnosis on a background of previous treatment. Of these all with a pathogenic variant chose bilateral mastectomy. CONCLUSION Timely BRCA gene testing influences surgeons' and patients' choice of surgery. It reassures women with a negative result and allows those with a positive result to take an active decision about the management of their future risk.
Collapse
Affiliation(s)
- Quratul Ain
- Royal Marsden Hospital NHS Foundation Trust, UK
| | | | - Miriam Mutebi
- Royal Marsden Hospital NHS Foundation Trust, UK; Aga Khan University Hospital, Parklands, Nairobi, Kenya
| | - Angela George
- Royal Marsden Hospital NHS Foundation Trust, UK; Institute of Cancer Research, UK
| | - Zoe Kemp
- Royal Marsden Hospital NHS Foundation Trust, UK
| | - Jennifer E Rusby
- Royal Marsden Hospital NHS Foundation Trust, UK; Institute of Cancer Research, UK.
| |
Collapse
|
32
|
Wu HL, Luo ZY, He ZL, Gong Y, Mo M, Ming WK, Liu GY. All HER2-negative breast cancer patients need gBRCA testing: cost-effectiveness and clinical benefits. Br J Cancer 2023; 128:638-646. [PMID: 36564566 PMCID: PMC9938252 DOI: 10.1038/s41416-022-02111-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The OlympiA trial demonstrated the benefits of adjuvant usage of olaparib for high-risk patients with human epidermal growth factor receptor 2 (HER2)-negative breast cancer (BC) and germline BRCA (gBRCA) mutation. This provoked thoughts on the clinical criteria of gBRCA testing. This study aims to estimate the costs and benefits of gBRCA testing and adjuvant olaparib therapy for patients with triple-negative breast cancer (TNBC) and hormone-receptor (HR)-positive and HER2-negative BC in China and the United States of America (USA). METHODS We used a Markov chain decision tree analytic model to compare three gBRCA screening policies in China and the USA: (1) no gBRCA testing; (2) selected gBRCA testing and (3) universal gBRCA testing for nonmetastatic TNBC and HR-positive HER2-negative BC patients. We modelled the benefit of systemic therapy and risk-reducing surgeries among patients identified with pathogenic or likely pathogenic variants (PVs) in BRCA1 and BRCA2. RESULTS Changing from the selected gBRCA testing to the universal gBRCA testing in TNBC patients is cost-effective, with the incremental cost-effectiveness ratios (ICERs) being 10991.1 and 56518.2 USD/QALY in China and the USA, respectively. Expanding universal gBRCA testing to HR-positive HER2-negative BC and TNBC patients has ICERs of 2023.3 and 16611.1 USD/QALY in China and the USA, respectively. DISCUSSION By performing gBRCA testing on all HER2-negative BC patients, adjuvant olaparib can be offered to high-risk patients with a PV in BRCA1 or BRCA2. These patients are also candidates for risk-reducing surgeries, an important aspect of their survivorship care, and these interventions can improve survival outcomes. With the willingness-to-pay thresholds being 31,500.0 and 100,000.0 USD per QALY gained in China and the USA, respectively, universal gBRCA testing is likely cost-effective for all HER2-negative BC patients. This simplified criterion of gBRCA testing for BC is recommended for adoption by current guidelines in China and the USA.
Collapse
Affiliation(s)
- Huai-Liang Wu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zi-Yin Luo
- Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Affiliated Hospital of Sun Yat‑sen University, Guangzhou, China
| | - Zong-Lin He
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Yue Gong
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Miao Mo
- Department of Cancer Prevention, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wai-Kit Ming
- Department of Infectious Diseases and Public Health, Jockey Club College of Veterinary Medicine and Life Science, City University of Hong Kong, Hong Kong SAR, China
| | - Guang-Yu Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Department of Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
| |
Collapse
|
33
|
Teppala S, Hodgkinson B, Hayes S, Scuffham P, Tuffaha H. A review of the cost-effectiveness of genetic testing for germline variants in familial cancer. J Med Econ 2023; 26:19-33. [PMID: 36426964 DOI: 10.1080/13696998.2022.2152233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Targeted germline testing is recommended for those with or at risk of breast, ovarian, or colorectal cancer. The affordability of genetic sequencing has improved over the past decade, therefore the cost-effectiveness of testing for these cancers is worthy of reassessment. OBJECTIVE To systematically review economic evaluations on cost-effectiveness of germline testing in breast, ovarian, or colorectal cancer. METHODS A search of PubMed and Embase databases for cost-effectiveness studies on germline testing in breast, ovarian, or colorectal cancer, published between 1999 and May 2022. Synthesis of methodology, cost-effectiveness, and reporting (CHEERS checklist) was performed. RESULTS The incremental cost-effectiveness ratios (ICERs; in 2021-adjusted US$) for germline testing versus the standard care option in hereditary breast or ovarian cancer (HBOC) across target settings were as follows: (1) population-wide testing: 344-2.5 million/QALY; (2) women with high-risk: dominant = 78,118/QALY, 8,337-59,708/LYG; (3) existing breast or ovarian cancer: 3,012-72,566/QALY, 39,835/LYG; and (4) metastatic breast cancer: 158,630/QALY. Likewise, ICERs of germline testing for colorectal cancer across settings were: (1) population-wide testing: 132,200/QALY, 1.1 million/LYG; (2) people with high-risk: 32,322-76,750/QALY, dominant = 353/LYG; and (3) patients with existing colorectal cancer: dominant = 54,122/QALY, 98,790-6.3 million/LYG. Key areas of underreporting were the inclusion of a health economic analysis plan (100% of HBOC and colorectal studies), engagement of patients and stakeholders (95.4% of HBOC, 100% of colorectal studies) and measurement of outcomes (18.2% HBOC, 38.9% of colorectal studies). CONCLUSION Germline testing for HBOC was likely to be cost-effective across most settings, except when used as a co-dependent technology with the PARP inhibitor, olaparib in metastatic breast cancer. In colorectal cancer studies, testing was cost-effective in those with high-risk, but inconclusive in other settings. Cost-effectiveness was sensitive to the prevalence of tested variants, cost of testing, uptake, and benefits of prophylactic measures. Policy advice on germline testing should emphasize the importance of these factors in their recommendations.
Collapse
Affiliation(s)
- Srinivas Teppala
- Centre for Applied Health Economics, Griffith University, Nathan, Australia
| | - Brent Hodgkinson
- Centre for Applied Health Economics, Griffith University, Nathan, Australia
| | - Sandi Hayes
- Menzies Health Institute Queensland, Griffith University, Southport, Australia
| | - Paul Scuffham
- Centre for Applied Health Economics, Griffith University, Nathan, Australia
- Menzies Health Institute Queensland, Griffith University, Southport, Australia
| | - Haitham Tuffaha
- Centre for the Business and Economics of Health, The University of Queensland, St. Lucia, Australia
| |
Collapse
|
34
|
Bell KA, Kim R, Aronson M, Gillies B, Ali Awan A, Chun K, Hart J, Healey R, Kim L, Klaric G, Panabaker K, Sabatini PJB, Sadikovic B, Selvarajah S, Smith AC, Stockley TL, Vaags AK, Eisen A, Pollett A, Feilotter H. Development of a comprehensive approach to adult hereditary cancer testing in Ontario. J Med Genet 2022:jmg-2022-108945. [PMID: 36564171 DOI: 10.1136/jmg-2022-108945] [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: 09/16/2022] [Accepted: 12/10/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Genetic testing for hereditary cancer susceptibility has advanced over time due to the discovery of new risk genes, improved technology and decreased cost. In the province of Ontario, testing eligibility criteria were initially developed to include hereditary breast, ovarian and colorectal cancer syndromes. The rapid evolution of genetic technologies has facilitated the ability to interrogate a large number of genes concurrently. This, coupled with new knowledge about risk genes, necessitated a coordinated approach to expanding the scope of genes and indications tested and synchronisation of access and test utilisation across the province as required in a publicly funded universal healthcare system. METHODS Ontario Health-Cancer Care Ontario convened expert working groups to develop a standardised and comprehensive cancer gene list for adults and accompanying hereditary cancer testing (HCT) criteria using an evidence-based framework and broad laboratory and clinical genetics engagement. RESULTS A standardised 76-cancer-gene panel, organised into 13 larger disease site panels and 25 single/small gene panels, was developed and endorsed by the working groups. Provincial genetic testing eligibility criteria were updated to align with the new panels and to guide clinical decision-making. In the first year following the implementation of these changes, 10 564 HCT panels were performed with an overall mutation detection rate of 12.2%. CONCLUSION Using an evidence framework and broad clinical engagement to develop and endorse an updated guidance document, cancer genetic testing for adults in Ontario is now standardised and coordinated across the province.
Collapse
Affiliation(s)
| | - Raymond Kim
- Division of Medical Oncology and Hematology, University Health Network, Toronto, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Melyssa Aronson
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Brittany Gillies
- Familial Cancer Clinic, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Arif Ali Awan
- Division of Medical Oncology, Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada
| | - Kathy Chun
- The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer Hart
- Pathology and Laboratory Medicine Program, Ontario Health, Toronto, Ontario, Canada
| | - Rachel Healey
- Pathology and Laboratory Medicine Program, Ontario Health, Toronto, Ontario, Canada
| | - Linda Kim
- Department of Laboratory Medicine and Genetics, Credit Valley Hospital Site, Mississauga, Ontario, Canada
| | - Goran Klaric
- Pathology and Laboratory Medicine Program, Ontario Health, Toronto, Ontario, Canada
| | - Karen Panabaker
- Medical Genetics Program of Southwestern Ontario, London Health Sciences Centre, London, Ontario, Canada
| | | | - Bekim Sadikovic
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada.,Verspeeten Clinical Genome Centre, Western University, London, Ontario, Canada
| | - Shamini Selvarajah
- Department of Clinical Laboratory Genetics, University Health Network, Toronto, Ontario, Canada
| | | | - Tracy L Stockley
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada
| | - Andrea K Vaags
- Laboratory Medicine and Genetics, Trillium Health Partners, Mississauga, Ontario, Canada
| | - Andrea Eisen
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Aaron Pollett
- Pathology and Laboratory Medicine Program, Ontario Health, Toronto, Ontario, Canada.,Division of Diagnostic Medical Genetics, Sinai Health System, Toronto, Ontario, Canada
| | - Harriet Feilotter
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
35
|
Frey MK, Ahsan MD, Badiner N, Lin J, Narayan P, Nitecki R, Rauh-Hain JA, Moss H, Fowlkes RK, Thomas C, Bergeron H, Christos P, Levi SR, Blank SV, Holcomb K, Cantillo E, Sharaf RN, Lipkin S, Offit K, Chapman-Davis E. What happens in the long term: Uptake of cancer surveillance and prevention strategies among at-risk relatives with pathogenic variants detected via cascade testing. Cancer 2022; 128:4241-4250. [PMID: 36305018 PMCID: PMC10041659 DOI: 10.1002/cncr.34482] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Cascade genetic testing for hereditary cancer syndromes offers affected relatives the opportunity to pursue cancer screening and risk-reducing surgery and thus reduces morbidity and mortality. The purpose of this study was to measure the long-term utilization of targeted cancer prevention and quality of life among at-risk relatives offered clinician-facilitated cascade genetic testing. METHODS In a pilot study, at-risk relatives of patients with a hereditary cancer syndrome were contacted directly by the clinical team and offered telephone genetic counseling and genetic testing via an at-home, mailed saliva kit. Two-year follow-up results evaluating the use of targeted cancer prevention strategies and the quality of life for enrolled relatives were reported. Quality-of-life was measured with validated surveys, and scores were compared to the time of initial contact by the Wilcoxon signed-rank test. RESULTS Ninety-five at-risk relatives were enrolled in the initial pilot study, and 72 (76%) participated in the 2-year follow-up; 57 of these (79%) had completed genetic testing. Twenty-five of those 57 relatives (44%) were found to harbor an inherited pathogenic variant. Guideline-based cancer surveillance was recommended to 18 relatives; 13 (72%) completed at least one recommended screening, and six (33%) completed all recommended screenings. Risk-reducing surgery was recommended to 10 relatives; four (40%) completed a total of eight procedures. Quality-of-life surveys demonstrated low levels of anxiety, depression, distress, and uncertainty. CONCLUSIONS The 2-year follow-up of the original pilot study revealed that clinician-facilitated cascade testing resulted in genetically targeted cancer screening and prevention with preserved quality of life. These results, to be confirmed by larger randomized controlled trials, suggest that medical systems should consider supporting clinician-facilitated cascade testing programs.
Collapse
Affiliation(s)
| | | | | | - Jenny Lin
- Weill Cornell Medicine, New York, New York, USA
| | | | - Roni Nitecki
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Haley Moss
- Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | | | | | | | | | | | | | | | - Kenneth Offit
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | |
Collapse
|
36
|
van der Merwe NC, Ntaita KS, Stofberg H, Combrink HM, Oosthuizen J, Kotze MJ. Implementation of multigene panel testing for breast and ovarian cancer in South Africa: A step towards excellence in oncology for the public sector. Front Oncol 2022; 12:938561. [PMID: 36568162 PMCID: PMC9768488 DOI: 10.3389/fonc.2022.938561] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 11/08/2022] [Indexed: 12/13/2022] Open
Abstract
Translation of genomic knowledge into public health benefits requires the implementation of evidence-based recommendations in clinical practice. In this study, we moved beyond BRCA1/2 susceptibility testing in breast and ovarian cancer patients to explore the application of pharmacogenetics across multiple genes participating in homologous recombination DNA damage repair. This involved the utilisation of next-generation sequencing (NGS) at the intersection of research and service delivery for development of a comprehensive genetic testing platform in South Africa. Lack of international consensus regarding risk categorization of established cancer susceptibility genes and the level of evidence required for prediction of drug response supported the development of a central database to facilitate clinical interpretation. Here we demonstrate the value of this approach using NGS to 1) determine the variant spectrum applicable to targeted therapy and implementation of prevention strategies using the 15-gene Oncomine™ BRCA Expanded Panel, and 2) searched for novel and known pathogenic variants in uninformative cases using whole exome sequencing (WES). Targeted NGS performed as a routine clinical service in 414 South African breast and/or ovarian cancer patients resulted in the detection of 48 actionable variants among 319 (15%) cases. BRCA1/2-associated cancers were identified in 70.8% of patients (34/48, including two double-heterozygotes), with the majority (35.3%, 12/34) representing known South African founder variants. Detection of actionable variants in established non-BRCA1/2 risk genes contributed 29% to the total percentage (14/48), distributed amongst ATM, CHEK2, BARD1, BRIP1, PALB2 and TP53. Experimental WES using a virtually constructed multi-cancer NGS panel in 16 genetically unresolved cases (and four controls) revealed novel protein truncating variants in the basal cell carcinoma gene PTCH1 (c.4187delG) and the signal transmission and transduction gene KIT (c.930delA) involved in crucial cellular processes. Based on these findings, the most cost-effective approach would be to perform BRCA1/2 founder variant testing at referral, followed by targeted multigene panel testing if clinically indicated and addition of WES in unresolved cases. This inventive step provides a constant flow of new knowledge into the diagnostic platform via a uniquely South African pathology-supported genetic approach implemented for the first time in this context to integrate research with service delivery.
Collapse
Affiliation(s)
- Nerina C. van der Merwe
- Division of Human Genetics, National Health Laboratory Service, Universitas Hospital, Bloemfontein, South Africa,Division of Human Genetics, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa,*Correspondence: Nerina C. van der Merwe,
| | - Kholiwe S. Ntaita
- Division of Human Genetics, National Health Laboratory Service, Universitas Hospital, Bloemfontein, South Africa,Division of Human Genetics, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Hanri Stofberg
- Division of Human Genetics, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Herkulaas MvE. Combrink
- Office of the Dean, Economic and Management Sciences, University of the Free State, Bloemfontein, South Africa,Interdisciplinary Centre for Digital Futures, University of the Free State, Bloemfontein, South Africa
| | - Jaco Oosthuizen
- Division of Human Genetics, National Health Laboratory Service, Universitas Hospital, Bloemfontein, South Africa,Division of Human Genetics, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Maritha J. Kotze
- Division of Chemical Pathology, Department of Pathology, National Health Laboratory Service, Tygerberg Hospital, Cape Town, South Africa,Division of Chemical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
37
|
Germline Genetic Testing in Breast Cancer: Systemic Therapy Implications. Curr Oncol Rep 2022; 24:1791-1800. [PMID: 36255604 DOI: 10.1007/s11912-022-01340-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW In this article, we discuss recent advances in germline genetic testing for patients with breast cancer and highlight current limitations and impacts on clinical care. We also provide an update on the therapeutic implications of having a germline mutation, including targeted systemic therapy options for treating early and metastatic breast cancer. RECENT FINDINGS Approximately 5 to 10% of women diagnosed with breast cancer have a pathogenic variant in a hereditary cancer susceptibility gene, which has significant implications for managing these patients. Previously, testing was done mainly to inform screening and risk-reduction treatment; however, more recently, germline genetic results have significant systemic therapy implications that can meaningfully improve outcomes in breast cancer patients, especially with oral poly-ADP-ribose polymerase (PARP) inhibitors. These systemic therapy advances implore a shift in paradigm for whom to test moving forward and how to modify the existing testing models to meet the increasing demand for germline testing, which is expected to grow exponentially.
Collapse
|
38
|
Muacevic A, Adler JR, Torres-Lopez D, Olaya L, Gutierrez-Vargas M, Olaya G, Olaya JD. Sociodemographic, Clinical, and Variation Outcomes for Breast Cancer and Breast Cancer-Related Mutations in a Ten-Year Cohort From Neiva, Huila, Colombia. Cureus 2022; 14:e32257. [PMID: 36620844 PMCID: PMC9815492 DOI: 10.7759/cureus.32257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2022] [Indexed: 12/12/2022] Open
Abstract
Background Some breast cancer cases are related to inherited mutations, and this is the reason why early mutation screening is emerging as an area of focus for cost-effective care. However, breast cancer-related mutations vary according to race, ethnicity, geographic origin, and healthcare access. Surveillance for familial breast cancer is not performed routinely in Colombia. Our main aim in this study was to describe a cohort of breast cancer patients, carrying founder breast cancer gene (BRCA) mutations, which were followed up for up to 10 years (2010-2019) in Neiva, Colombia. Methods We performed a retrospective description from an outpatient care center in Huila, Colombia, a region with high breast cancer rates. This study included patients with both a breast cancer diagnosis and an incident genetic mutation for breast cancer (detected during a breast cancer consultation). We captured information from patient medical records. Descriptive analyses were performed. Results A total of 105 patients met the study's inclusion criteria and were included patients with the BRCA1 mutation and three with BRCA2 mutations. They had a median age of 45 years (IQR, 36 to 51 years). Relatives with a breast cancer history were found in 74 carriers (70.5%). Most patients had a report of Breast Imaging-Reporting and Data System (BIRADS) ≥ 4. A TNM (tumor, node, metastasis) changed reclassification was observed in anatomical vs. prognostic classification. Median follow-up was of 74 months (IQR, 44 to 130), overall observed mortality was 22.9%, and specific mortality was 19.1%. Conclusion Women with breast cancer who carry a mutation related to breast cancer are usually younger than age 50 at diagnosis. Developing strategies and specific policies for this population is needed, and a prevalent BRCA1 c.3331_3334delCAAG mutation could be used as a cost-effective first approach. Among these patients, a risk-increased reclassification was observed.
Collapse
|
39
|
Torr B, Jones C, Choi S, Allen S, Kavanaugh G, Hamill M, Garrett A, MacMahon S, Loong L, Reay A, Yuan L, Valganon Petrizan M, Monson K, Perry N, Fallowfield L, Jenkins V, Gold R, Taylor A, Gabe R, Wiggins J, Lucassen A, Manchanda R, Gandhi A, George A, Hubank M, Kemp Z, Evans DG, Bremner S, Turnbull C. A digital pathway for genetic testing in UK NHS patients with cancer: BRCA-DIRECT randomised study internal pilot. J Med Genet 2022; 59:1179-1188. [PMID: 35868849 PMCID: PMC9691828 DOI: 10.1136/jmg-2022-108655] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/03/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Germline genetic testing affords multiple opportunities for women with breast cancer, however, current UK NHS models for delivery of germline genetic testing are clinician-intensive and only a minority of breast cancer cases access testing. METHODS We designed a rapid, digital pathway, supported by a genetics specialist hotline, for delivery of germline testing of BRCA1/BRCA2/PALB2 (BRCA-testing), integrated into routine UK NHS breast cancer care. We piloted the pathway, as part of the larger BRCA-DIRECT study, in 130 unselected patients with breast cancer and gathered preliminary data from a randomised comparison of delivery of pretest information digitally (fully digital pathway) or via telephone consultation with a genetics professional (partially digital pathway). RESULTS Uptake of genetic testing was 98.4%, with good satisfaction reported for both the fully and partially digital pathways. Similar outcomes were observed in both arms regarding patient knowledge score and anxiety, with <5% of patients contacting the genetics specialist hotline. All progression criteria established for continuation of the study were met. CONCLUSION Pilot data indicate preliminary demonstration of feasibility and acceptability of a fully digital pathway for BRCA-testing and support proceeding to a full powered study for evaluation of non-inferiority of the fully digital pathway, detailed quantitative assessment of outcomes and operational economic analyses. TRIAL REGISTRATION NUMBER ISRCTN87845055.
Collapse
Affiliation(s)
- Bethany Torr
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
| | - Christopher Jones
- Clinical Trials Unit, Brighton and Sussex Medical School, Brighton, UK
| | - Subin Choi
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
| | - Sophie Allen
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
| | - Grace Kavanaugh
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
| | - Monica Hamill
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
| | - Alice Garrett
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
| | - Suzanne MacMahon
- Centre for Molecular Pathology, Institute of Cancer Research Sutton, Sutton, UK
| | - Lucy Loong
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
| | - Alistair Reay
- Centre for Molecular Pathology, Institute of Cancer Research Sutton, Sutton, UK
| | - Lina Yuan
- Centre for Molecular Pathology, Institute of Cancer Research Sutton, Sutton, UK
| | | | - Kathryn Monson
- Sussex Health Outcomes, Research and Education in Cancer (SHORE-C), Brighton and Sussex Medical School, Brighton, UK
| | - Nicky Perry
- Clinical Trials Unit, Brighton and Sussex Medical School, Brighton, UK
| | - Lesley Fallowfield
- Sussex Health Outcomes, Research and Education in Cancer (SHORE-C), Brighton and Sussex Medical School, Brighton, UK
| | - Valerie Jenkins
- Sussex Health Outcomes, Research and Education in Cancer (SHORE-C), Brighton and Sussex Medical School, Brighton, UK
| | | | - Amy Taylor
- Clinical Genetics, East Anglian Medical Genetics Service, Cambridge, UK
| | - Rhian Gabe
- Wolfson Institute of Population Health, Queen Mary's University of London, London, UK
| | - Jennifer Wiggins
- Cancer Genetics Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Anneke Lucassen
- Clinical Ethics and Law at Southampton (CELS), University of Southampton, Southampton, UK
- Department of Medicine, Univerity of Oxford Nuffield, Oxford, UK
| | - Ranjit Manchanda
- Wolfson Institute of Population Health, Queen Mary's University of London, London, UK
- Department of Gynaecological Oncology, Barts Health NHS Trust, London, UK
- Department of Health Services Research, Faculty of Public Health & Policy, London School of Hygiene and Tropical Medicine, London, UK
| | - Ashu Gandhi
- School of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Prevent Breast Cancer Centre, Wythenshawe Hospital Manchester Universities Foundation Trust, Manchester, UK
| | - Angela George
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
- Cancer Genetics Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - Michael Hubank
- Centre for Molecular Pathology, Institute of Cancer Research Sutton, Sutton, UK
| | - Zoe Kemp
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
- Cancer Genetics Unit, Royal Marsden NHS Foundation Trust, London, UK
| | - D Gareth Evans
- Nightingale and Genesis Breast Cancer Centre, University Hospital of South Manchester NHS Foundation Trust, Manchester, UK
- Division of Evolution and Genomic Sciences, The University of Manchester, Manchester, UK
| | - Stephen Bremner
- Clinical Trials Unit, Brighton and Sussex Medical School, Brighton, UK
| | - Clare Turnbull
- Institute of Cancer Research, Division of Genetics and Epidemiology, Sutton, UK
- Cancer Genetics Unit, Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
40
|
Leite ACR, Suzuki DA, Pereira AAL, Machado NP, Barroso-Sousa R, Correa TS, Moura FC, Morbeck IAP, Gumz BP, Faria LDBB, Fernandes GDS, Sandoval RL. What can we learn from more than 1,000 Brazilian patients at risk of hereditary cancer? Front Oncol 2022; 12:963910. [PMID: 36132150 PMCID: PMC9484549 DOI: 10.3389/fonc.2022.963910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Identifying individuals at a higher risk of developing cancer is a major concern for healthcare providers. Cancer predisposition syndromes are the underlying cause of cancer aggregation and young-onset tumors in many families. Germline genetic testing is underused due to lack of access, but Brazilian germline data associated with cancer predisposition syndromes are needed. Methods Medical records of patients referred for genetic counseling at the Oncogenetics Department at the Hospital Sírio-Libanês (Brasília, DF, Brazil) from July 2017 to January 2021 were reviewed. The clinical features and germline findings were described. Detection rates of germline pathogenic/likely pathogenic variant (P/LPV) carriers were compared between international and Brazilian guidelines for genetic testing. Results A total of 1,091 individuals from 985 families were included in this study. Most patients (93.5%) had a family history of cancer, including 64% with a family member under 50 with cancer. Sixty-six percent of patients (720/1091) had a personal history of cancer. Young-onset cancers (<50 years old) represented 62% of the patients affected by cancer and 17% had multiple primary cancers. The cohort included patients with 30 different cancer types. Breast cancer was the most prevalent type of cancer (52.6%). Germline testing included multigene panel (89.3%) and family variant testing (8.9%). Approximately 27% (236/879) of the tested patients harbored germline P/LPVs in cancer susceptibility genes. BRCA2, BRCA1, and TP53 were the most frequently reported genes, corresponding to 18.6%, 14.4%, and 13.5% of the positive results, respectively. Genetic testing criteria from international guidelines were more effective in identifying carriers than the Brazilian National Agency of Supplementary Health (ANS) criteria (92% vs. 72%, p<0.001). Forty-six percent of the cancer-unaffected patients who harbored a germline P/LPV (45/98) would not be eligible for genetic testing according to ANS because they did not have a family variant previously identified in a cancer-affected relative. Conclusion The high detection rate of P/LPVs in the present study is possibly related to the genetic testing approach with multigene panels and cohort's characteristics, represented mainly by individuals with a personal or family history of young-onset cancer. Testing asymptomatic individuals with suspicious family history may also have contributed to a higher detection rate. A significant number of carriers would not have been identified using ANS criteria for genetic testing.
Collapse
|
41
|
Manchanda R, Sideris M. Population based genetic testing for cancer susceptibility genes: quo vadis. BJOG 2022; 130:125-130. [PMID: 36017754 PMCID: PMC10087260 DOI: 10.1111/1471-0528.17283] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/23/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Ranjit Manchanda
- Wolfson Institute of Population Health, Barts CRUK Cancer Centre, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.,Department of Gynaecological Oncology, Barts Health NH Trust, EC1A 7BE, London, UK.,Department of Health Services Research and Policy, School of Hygiene & Tropical Medicine, London WC1H 9SH, London, UK.,Department of Gynaecology, All India Institute of Medical Sciences, New Delhi, India
| | - Michail Sideris
- Wolfson Institute of Population Health, Barts CRUK Cancer Centre, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.,Department of Gynaecological Oncology, Barts Health NH Trust, EC1A 7BE, London, UK
| |
Collapse
|
42
|
Sun L, Wei X, Manchanda R, Legood R, Yang L. Using Genetic Testing at Cancer Diagnosis for Breast Cancer Control. China CDC Wkly 2022; 4:711-714. [PMID: 36060444 PMCID: PMC9433755 DOI: 10.46234/ccdcw2022.148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/09/2022] [Indexed: 12/02/2022] Open
Affiliation(s)
- Li Sun
- Department of Health Services Research and Policy, Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, London, UK.,Wolfson Institute of Population Health, Barts CRUK Cancer Centre, Queen Mary University of London, London, UK
| | - Xia Wei
- Department of Health Services Research and Policy, Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, London, UK.,Wolfson Institute of Population Health, Barts CRUK Cancer Centre, Queen Mary University of London, London, UK
| | - Ranjit Manchanda
- Department of Health Services Research and Policy, Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, London, UK.,Wolfson Institute of Population Health, Barts CRUK Cancer Centre, Queen Mary University of London, London, UK.,Department of Gynaecological Oncology, Barts Health NHS Trust, London, UK
| | - Rosa Legood
- Department of Health Services Research and Policy, Faculty of Public Health and Policy, London School of Hygiene and Tropical Medicine, London, UK
| | - Li Yang
- School of Public Health, Peking University, Beijing, China
| |
Collapse
|
43
|
Wang L, Zhang YL, Jiang C, Duan FF, Yuan ZY, Huang JJ, Bi XW. Novel Signatures Based on the Lymphocyte-to-C-Reactive Protein Ratio Predict the Prognosis of Patients with Early Breast Cancer: A Retrospective Study. J Inflamm Res 2022; 15:3957-3974. [PMID: 35860229 PMCID: PMC9289276 DOI: 10.2147/jir.s364284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/07/2022] [Indexed: 01/08/2023] Open
Abstract
Background The value of the lymphocyte-to-C-reactive protein (CRP) ratio (LCR) in early breast cancer (BC) is unclear. We explored the correlation between the LCR and survival of patients with early BC and established effective LCR-based prognostic signatures for predicting prognosis. Methods In this retrospective study, we randomized 623 patients with early-stage BC diagnosed in December 2010 to October 2012 at the Sun Yat-sen University Cancer Center to training and verification datasets. The median follow-up of all patients was 109 months. The survival differences were calculated by Kaplan–Meier method using the Log rank test. For overall survival (OS) and disease-free survival (DFS), the independent factors in the training dataset were identified using univariate and multivariate Cox analyses, in which two-tailed P-values < 0.05 were considered statistically significant. Based on this, we respectively constructed novel signatures for survival prediction and validated the efficiency of signatures through the concordance index (C-index), calibration and receiver operating characteristic (ROC) curves in both datasets. Results The LCR, lymphatic vessel invasion (LVI), progesterone receptor (PR) status, and Ki67 index were independent prognostic factors of OS. And the LCR and LVI are associated to DFS too. High LCR was associated with better OS and DFS. We constructed the prediction signatures based on those independent prognostic factors and calculated the risk scores. Patients in the training dataset with higher risk scores had significantly worse prognosis (P < 0.001). The signature had excellent discrimination capacity, with an OS C-index of 0.785 [95% confidence interval (CI): 0.713–0.857] and 0.750 (95% CI: 0.669–0.832) in the training and verification datasets, respectively. The time–ROC curves also suggest accurate prediction by the signature. Conclusion The LCR was a significant prognostic predictor of OS and DFS in early BC. The LCR-based prognostic signatures could be a useful tool for individualized therapeutic guidance.
Collapse
Affiliation(s)
- Li Wang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yu-Ling Zhang
- Department of Endocrinology, Jiangxi Provincial People's Hospital, Nanchang, People's Republic of China
| | - Chang Jiang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Fang-Fang Duan
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Zhong-Yu Yuan
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jia-Jia Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Xi-Wen Bi
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| |
Collapse
|
44
|
Lourenção M, Simões Correa Galendi J, Galvão HDCR, Antoniazzi AP, Grasel RS, Carvalho AL, Mauad EC, de Oliveira JHC, Reis RM, Mandrik O, Palmero EI. Cost-Effectiveness of BRCA 1/2 Genetic Test and Preventive Strategies: Using Real-World Data From an Upper-Middle Income Country. Front Oncol 2022; 12:951310. [PMID: 35898894 PMCID: PMC9309566 DOI: 10.3389/fonc.2022.951310] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Although BRCA1/2 genetic testing in developed countries is part of the reality for high-risk patients for hereditary breast and ovarian cancer (HBOC), the same is not true for upper-middle-income countries. For that reason, this study aimed to evaluate whether the BRCA1/2 genetic test and preventive strategies for women at high risk for HBOC are cost-effective compared to not performing these strategies in an upper-middle-income country. Adopting a payer perspective, a Markov model with a time horizon of 70 years was built to delineate the health states for a cohort of healthy women aged 30 years that fulfilled the BRCA1/2 testing criteria according to the guidelines. Transition probabilities were calculated based on real-world data of women tested for BRCA1/2 germline mutations in a cancer reference hospital from 2011 to 2020. We analyzed 275 BRCA mutated index cases and 356 BRCA mutation carriers that were first- or second-degree relatives of the patients. Costs were based on the Brazilian public health system reimbursement values. Health state utilities were retrieved from literature. The BRCA1/2 genetic test and preventive strategies result in more quality-adjusted life years (QALYs) and costs with an incremental cost-effectiveness ratio of R$ 11,900.31 (U$ 5,504.31)/QALY. This result can represent a strong argument in favor of implementing genetic testing strategies for high-risk women even in countries with upper-middle income, considering not only the cancer prevention possibilities associated with the genetic testing but also its cost-effectiveness to the health system. These strategies are cost-effective, considering a willingness-to-pay threshold of R$ 25,000 (U$ 11,563.37)/QALY, indicating that the government should consider offering them for women at high risk for HBOC. The results were robust in deterministic and probabilistic sensitivity analyses.
Collapse
Affiliation(s)
- Marina Lourenção
- School of Economics, Business Administration and Accounting at Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - Julia Simões Correa Galendi
- Institute of Health Economics and Clinical Epidemiology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | | | | | - Rebeca Silveira Grasel
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
- Department of Genetics, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - André Lopes Carvalho
- Early Detection Prevention and Infections, International Agency for Research on Cancer, Lyon, France
| | | | | | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Guimarães, Portugal
| | - Olena Mandrik
- School of Health and Related Research, The University of Sheffield, Sheffield, United Kingdom
| | - Edenir Inêz Palmero
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
- Department of Genetics, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| |
Collapse
|
45
|
Lu T, Forgetta V, Richards JB, Greenwood CMT. Polygenic risk score as a possible tool for identifying familial monogenic causes of complex diseases. Genet Med 2022; 24:1545-1555. [PMID: 35460399 DOI: 10.1016/j.gim.2022.03.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 01/13/2023] Open
Abstract
PURPOSE The study aimed to evaluate whether polygenic risk scores could be helpful in addition to family history for triaging individuals to undergo deep-depth diagnostic sequencing for identifying monogenic causes of complex diseases. METHODS Among 44,550 exome-sequenced European ancestry UK Biobank participants, we identified individuals with a clinically reported or computationally predicted monogenic pathogenic variant for breast cancer, bowel cancer, heart disease, diabetes, or Alzheimer disease. We derived polygenic risk scores for these diseases. We tested whether a polygenic risk score could identify rare pathogenic variant heterozygotes among individuals with a parental disease history. RESULTS Monogenic causes of complex diseases were more prevalent among individuals with a parental disease history than in the rest of the population. Polygenic risk scores showed moderate discriminative power to identify familial monogenic causes. For instance, we showed that prescreening the patients with a polygenic risk score for type 2 diabetes can prioritize individuals to undergo diagnostic sequencing for monogenic diabetes variants and reduce needs for such sequencing by up to 37%. CONCLUSION Among individuals with a family history of complex diseases, those with a low polygenic risk score are more likely to have monogenic causes of the disease and could be prioritized to undergo genetic testing.
Collapse
Affiliation(s)
- Tianyuan Lu
- Centre for Clinical Epidemiology, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Quantitative Life Sciences Program, McGill University, Montreal, Quebec, Canada
| | - Vincenzo Forgetta
- Centre for Clinical Epidemiology, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - John Brent Richards
- Centre for Clinical Epidemiology, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada; Department of Twin Research and Genetic Epidemiology, School of Life Course & Population Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Celia M T Greenwood
- Centre for Clinical Epidemiology, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada; Department of Epidemiology, Biostatistics and Occupational Health, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, Gerald Bronfman Department of Oncology, McGill University, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
46
|
Gynecologic Cancer Risk and Genetics: Informing an Ideal Model of Gynecologic Cancer Prevention. Curr Oncol 2022; 29:4632-4646. [PMID: 35877228 PMCID: PMC9322111 DOI: 10.3390/curroncol29070368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/09/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022] Open
Abstract
Individuals with proven hereditary cancer syndrome (HCS) such as BRCA1 and BRCA2 have elevated rates of ovarian, breast, and other cancers. If these high-risk people can be identified before a cancer is diagnosed, risk-reducing interventions are highly effective and can be lifesaving. Despite this evidence, the vast majority of Canadians with HCS are unaware of their risk. In response to this unmet opportunity for prevention, the British Columbia Gynecologic Cancer Initiative convened a research summit “Gynecologic Cancer Prevention: Thinking Big, Thinking Differently” in Vancouver, Canada on 26 November 2021. The aim of the conference was to explore how hereditary cancer prevention via population-based genetic testing could decrease morbidity and mortality from gynecologic cancer. The summit invited local, national, and international experts to (1) discuss how genetic testing could be more broadly implemented in a Canadian system, (2) identify key research priorities in this topic and (3) outline the core essential elements required for such a program to be successful. This report summarizes the findings from this research summit, describes the current state of hereditary genetic programs in Canada, and outlines incremental steps that can be taken to improve prevention for high-risk Canadians now while developing an organized population-based hereditary cancer strategy.
Collapse
|
47
|
Zischke J, White N, Gordon L. Accounting for Intergenerational Cascade Testing in Economic Evaluations of Clinical Genomics: A Scoping Review. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2022; 25:944-953. [PMID: 35667782 DOI: 10.1016/j.jval.2021.11.1353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 10/25/2021] [Accepted: 11/03/2021] [Indexed: 06/15/2023]
Abstract
OBJECTIVES Clinical genomics is emerging as a diagnostic tool in the identification of blood relatives at risk of developing heritable diseases. Our objective was to identify how genetic cascade screening has been incorporated into health economic evaluations. METHODS A scoping review was conducted to identify how multiple generations of a family were included in economic evaluations of clinical genomic sequencing, how many and which relatives were included, and uptake rates. Databases were searched for full economic evaluations of genetic interventions that screened multiple generations of families and were in English language, and no restrictions were made for disease or publication type. Data were synthesized using a narrative approach. RESULTS Twenty-five studies were included covering a range of diseases in various countries. Markov cohort models were mostly used with hypothetical populations and unsupported by clinical evidence. Cascade testing was either the primary intervention or secondary to the index cases. The number and type of relatives were based on assumptions or identified through population or family records, clinical registry data, or clinical literature. Studies included only immediate family members and the uptake of testing ranged between 20% and 100%. All interventions were reported as cost-effective, and a higher number of relatives was a key driver. CONCLUSIONS Several economic evaluations have considered the impacts of cascade testing interventions within clinical genomics. Ideally, models supported with high-quality clinical data are needed and, in their absence, transparent and justifiable assumptions of uptake rates and choices about including relatives. Consideration of more appropriate modeling types is required.
Collapse
Affiliation(s)
- Jason Zischke
- Health Economics Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia; School of Public Health and Social Work, Queensland University of Technology, Brisbane, Australia.
| | - Nicole White
- Centre for Healthcare Transformation, School of Public Health and Social Work and Australian Centre for Health Services Innovation, Queensland University of Technology, Brisbane, Australia
| | - Louisa Gordon
- Health Economics Group, QIMR Berghofer Medical Research Institute, Brisbane, Australia; School of Nursing, Queensland University of Technology, Brisbane, Australia; School of Public Health, The University of Queensland, Brisbane, Australia
| |
Collapse
|
48
|
Mital S, Nguyen HV. Cost-effectiveness of using artificial intelligence versus polygenic risk score to guide breast cancer screening. BMC Cancer 2022; 22:501. [PMID: 35524200 PMCID: PMC9074290 DOI: 10.1186/s12885-022-09613-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/26/2022] [Indexed: 11/10/2022] Open
Abstract
Background Current guidelines for mammography screening for breast cancer vary across agencies, especially for women aged 40–49. Using artificial Intelligence (AI) to read mammography images has been shown to predict breast cancer risk with higher accuracy than alternative approaches including polygenic risk scores (PRS), raising the question whether AI-based screening is more cost-effective than screening based on PRS or existing guidelines. This study provides the first evidence to shed light on this important question. Methods This study is a model-based economic evaluation. We used a hybrid decision tree/microsimulation model to compare the cost-effectiveness of eight strategies of mammography screening for women aged 40–49 (screening beyond age 50 follows existing guidelines). Six of these strategies were defined by combinations of risk prediction approaches (AI, PRS or family history) and screening frequency for low-risk women (no screening or biennial screening). The other two strategies involved annual screening for all women and no screening, respectively. Data used to populate the model were sourced from the published literature. Results Risk prediction using AI followed by no screening for low-risk women is the most cost-effective strategy. It dominates (i.e., costs more and generates fewer quality adjusted life years (QALYs)) strategies for risk prediction using PRS followed by no screening or biennial screening for low-risk women, risk prediction using AI or family history followed by biennial screening for low-risk women, and annual screening for all women. It also extendedly dominates (i.e., achieves higher QALYs at a lower incremental cost per QALY) the strategy for risk prediction using family history followed by no screening for low-risk women. Meanwhile, it is cost-effective versus no screening, with an incremental cost-effectiveness ratio of $23,755 per QALY gained. Conclusions Risk prediction using AI followed by no breast cancer screening for low-risk women is the most cost-effective strategy. This finding can be explained by AI’s ability to identify high-risk women more accurately than PRS and family history (which reduces the possibility of delayed breast cancer diagnosis) and fewer false-positive diagnoses from not screening low-risk women. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09613-1.
Collapse
Affiliation(s)
- Shweta Mital
- School of Pharmacy, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John's, NL A1B 3V6, Canada
| | - Hai V Nguyen
- School of Pharmacy, Memorial University of Newfoundland, 300 Prince Philip Drive, St. John's, NL A1B 3V6, Canada.
| |
Collapse
|
49
|
Sun L, Cui B, Wei X, Sadique Z, Yang L, Manchanda R, Legood R. Cost-Effectiveness of Genetic Testing for All Women Diagnosed with Breast Cancer in China. Cancers (Basel) 2022; 14:cancers14071839. [PMID: 35406611 PMCID: PMC8997428 DOI: 10.3390/cancers14071839] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Unselected multigene testing at breast cancer (BC) diagnosis has been reported to be cost-effective compared with family history (FH)/clinical-criteria-based testing in high-income countries such as the US and UK. Chinese patients are younger than Caucasian women at diagnosis, tending to have a higher gene mutation prevalence, and the family size and number of female relatives are smaller due to the one-child policy (which has been changed) in China. Therefore, offering genetic testing for BC patients could potentially prevent more cancer cases and deaths in China. However, the health economic evidence for multigene testing at BC diagnosis in China is lacking. The aim of the current study was to evaluate the cost-effectiveness of three genetic testing strategies among BC patients using a microsimulation model at the individual level in China. We found that offering unselected multigene testing to all BC patients in China is highly cost-effective compared with FH/clinical-criteria-based testing or no testing from both the societal and payer perspectives. Abstract Unselected multigene testing for all women with breast cancer (BC) identifies more cancer susceptibility gene (CSG) carriers who can benefit from precision prevention compared with family history (FH)/clinical-criteria-based guidelines. Very little CSG testing is undertaken in middle-income countries such as China, and its cost-effectiveness remains unaddressed. We aimed to estimate cost-effectiveness and population impact of multigene testing for all Chinese BC patients. Data from 8085 unselected BC patients recruited to a Peking University Cancer Hospital study were used for microsimulation modeling, comparing three strategies in the Chinese setting: all BC women undergo BRCA1/BRCA2/PALB2 genetic testing, only BC women fulfilling FH/clinical criteria undergo BRCA testing, and no genetic testing. Prophylactic mastectomy and salpingo-oophorectomy would be adopted where appropriate. Societal and payer perspectives with a lifetime horizon along with sensitivity analyses were presented. Incremental cost-effectiveness ratio (ICER): incremental cost per quality-adjusted life-year (QALY) gained is compared to the USD 10,260/QALY (one-times GDP per capita) willingness-to-pay threshold. BC incidence, ovarian cancer (OC) incidence, and related deaths were also estimated. FH/clinical-criteria-based BRCA testing was ruled out on the principle of extensive dominance. Compared with no genetic testing, multigene testing for all BC patients had an ICER = USD 4506/QALY (societal perspective) and USD 7266/QALY (payer perspective), well below our threshold. Probabilistic sensitivity analysis showed unselected multigene testing remained cost-effective for 94.2%/86.6% of simulations from the societal and payer perspectives. One year’s unselected multigene testing could prevent 7868 BC/OC cases and 5164 BC/OC deaths in China. Therefore, unselected multigene testing is extremely cost-effective and should be offered to all Chinese women with BC.
Collapse
Affiliation(s)
- Li Sun
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London WC1H 9SH, UK
- Wolfson Institute for Population Health, CRUK Barts Cancer Centre, Queen Mary University of London, London EC1M 6BQ, UK
| | - Bin Cui
- School of Public Health, Peking University, Beijing 100191, China
| | - Xia Wei
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London WC1H 9SH, UK
- Wolfson Institute for Population Health, CRUK Barts Cancer Centre, Queen Mary University of London, London EC1M 6BQ, UK
- School of Public Health, Peking University, Beijing 100191, China
| | - Zia Sadique
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London WC1H 9SH, UK
| | - Li Yang
- School of Public Health, Peking University, Beijing 100191, China
| | - Ranjit Manchanda
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London WC1H 9SH, UK
- Wolfson Institute for Population Health, CRUK Barts Cancer Centre, Queen Mary University of London, London EC1M 6BQ, UK
- Department of Gynaecological Oncology, Barts Health NHS Trust, Royal London Hospital, London E1 1BB, UK
- Department of Gynaecology, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Rosa Legood
- Department of Health Services Research and Policy, London School of Hygiene & Tropical Medicine, London WC1H 9SH, UK
| |
Collapse
|
50
|
Simões Corrêa Galendi J, Kautz-Freimuth S, Stock S, Müller D. Uptake Rates of Risk-Reducing Surgeries for Women at Increased Risk of Hereditary Breast and Ovarian Cancer Applied to Cost-Effectiveness Analyses: A Scoping Systematic Review. Cancers (Basel) 2022; 14:1786. [PMID: 35406563 PMCID: PMC8997187 DOI: 10.3390/cancers14071786] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/23/2022] [Accepted: 03/26/2022] [Indexed: 01/09/2023] Open
Abstract
The cost-effectiveness of genetic screen-and-treat strategies for women at increased risk for breast and ovarian cancer often depends on the women's willingness to make use of risk-reducing mastectomy (RRM) or salpingo-oophorectomy (RRSO). To explore the uptake rates of RRM and RRSO applied in health economic modeling studies and the impact of uptake rates on the incremental cost-effectiveness ratios (ICER), we conducted a scoping literature review. In addition, using our own model, we conducted a value of information (VOI) analysis. Among the 19 models included in the review, the uptake rates of RRM ranged from 6% to 47% (RRSO: 10% to 88%). Fifty-seven percent of the models applied retrospective data obtained from registries, hospital records, or questionnaires. According to the models' deterministic sensitivity analyses, there is a clear trend that a lower uptake rate increased the ICER and vice versa. Our VOI analysis showed high decision uncertainty associated with the uptake rates. In the future, uptake rates should be given more attention in the conceptualization of health economic modeling studies. Prospective studies are recommended to reflect regional and national variations in women's preferences for preventive surgery.
Collapse
Affiliation(s)
- Julia Simões Corrêa Galendi
- Institute of Health Economics and Clinical Epidemiology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50935 Cologne, Germany; (S.K.-F.); (S.S.)
| | | | | | - Dirk Müller
- Institute of Health Economics and Clinical Epidemiology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, 50935 Cologne, Germany; (S.K.-F.); (S.S.)
| |
Collapse
|