1
|
Hasner MC, van Opijnen MP, de Vos FYF, Cuppen E, Broekman MLD. Whole genome sequencing in (recurrent) glioblastoma: challenges related to informed consent procedures and data sharing. Acta Neurochir (Wien) 2024; 166:266. [PMID: 38874628 PMCID: PMC11178618 DOI: 10.1007/s00701-024-06158-z] [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: 03/18/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024]
Abstract
Increased use of whole genome sequencing (WGS) in neuro-oncology for diagnostics and research purposes necessitates a renewed conversation about informed consent procedures and governance structures for sharing personal health data. There is currently no consensus on how to obtain informed consent for WGS in this population. In this narrative review, we analyze the formats and contents of frameworks suggested in literature for WGS in oncology and assess their benefits and limitations. We discuss applicability, specific challenges, and legal context for patients with (recurrent) glioblastoma. This population is characterized by the rarity of the disease, extremely limited prognosis, and the correlation of the stage of the disease with cognitive abilities. Since this has implications for the informed consent procedure for WGS, we suggest that the content of informed consent should be tailor-made for (recurrent) glioblastoma patients.
Collapse
Affiliation(s)
- Mira C Hasner
- Department of Neurosurgery, Haaglanden Medical Center, The Hague, The Netherlands
| | - Mark P van Opijnen
- Department of Neurosurgery, Haaglanden Medical Center, The Hague, The Netherlands.
- Department of Cell and Chemical Biology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands.
| | - Filip Y F de Vos
- Department of Medical Oncology, Utrecht University Medical Center, Utrecht, The Netherlands
| | - Edwin Cuppen
- Hartwig Medical Foundation, Amsterdam, The Netherlands
- Center for Molecular Medicine and Oncode Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marike L D Broekman
- Department of Neurosurgery, Haaglanden Medical Center, The Hague, The Netherlands
- Department of Cell and Chemical Biology, Leiden University Medical Center, Albinusdreef 2, Leiden, 2333 ZA, The Netherlands
- Department of Neurosurgery, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
Zhang H, Hussin H, Hoh CC, Cheong SH, Lee WK, Yahaya BH. Big data in breast cancer: Towards precision treatment. Digit Health 2024; 10:20552076241293695. [PMID: 39502482 PMCID: PMC11536614 DOI: 10.1177/20552076241293695] [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: 06/14/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Breast cancer is the most prevalent and deadliest cancer among women globally, representing a major threat to public health. In response, the World Health Organization has established the Global Breast Cancer Initiative framework to reduce breast cancer mortality through global collaboration. The integration of big data analytics (BDA) and precision medicine has transformed our understanding of breast cancer's biological traits and treatment responses. By harnessing large-scale datasets - encompassing genetic, clinical, and environmental data - BDA has enhanced strategies for breast cancer prevention, diagnosis, and treatment, driving the advancement of precision oncology and personalised care. Despite the increasing importance of big data in breast cancer research, comprehensive studies remain sparse, underscoring the need for more systematic investigation. This review evaluates the contributions of big data to breast cancer precision medicine while addressing the associated opportunities and challenges. Through the application of big data, we aim to deepen insights into breast cancer pathogenesis, optimise therapeutic approaches, improve patient outcomes, and ultimately contribute to better survival rates and quality of life. This review seeks to provide a foundation for future research in breast cancer prevention, treatment, and management.
Collapse
Affiliation(s)
- Hao Zhang
- Breast Cancer Translational Research Program (BCTRP@IPPT), Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
- Department of Biomedical Sciences, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| | - Hasmah Hussin
- Breast Cancer Translational Research Program (BCTRP@IPPT), Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
- Department of Clinical Medicine, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| | | | | | - Wei-Kang Lee
- Codon Genomics Sdn Bhd, Seri Kembangan, Selangor, Malaysia
| | - Badrul Hisham Yahaya
- Breast Cancer Translational Research Program (BCTRP@IPPT), Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
- Department of Biomedical Sciences, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Kepala Batas, Penang, Malaysia
| |
Collapse
|
3
|
Lam TC, Cho WCS, Au JSK, Ma ESK, Lam STS, Loong HHF, Wong JWH, Wong SM, Lee VHF, Leung RCY, Lau JKS, Kam MTY, Mok FST, Lim FMY, Nyaw JSF, Tin WWY, Cheung KM, Chan OSH, Kwong PWK, Cheung FY, Poon DM, Chik JYK, Lam MHC, Chan LWC, Wong SCC, Cao YB, Hui CV, Chen JZJ, Chang JH, Kong SFM, El Helali A. Consensus Statements on Precision Oncology in the China Greater Bay Area. JCO Precis Oncol 2023; 7:e2200649. [PMID: 37315266 PMCID: PMC10309548 DOI: 10.1200/po.22.00649] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/31/2023] [Accepted: 04/19/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Next-generation sequencing comprehensive genomic panels (NGS CGPs) have enabled the delivery of tailor-made therapeutic approaches to improve survival outcomes in patients with cancer. Within the China Greater Bay Area (GBA), territorial differences in clinical practices and health care systems and strengthening collaboration warrant a regional consensus to consolidate the development and integration of precision oncology (PO). Therefore, the Precision Oncology Working Group (POWG) formulated standardized principles for the clinical application of molecular profiling, interpretation of genomic alterations, and alignment of actionable mutations with sequence-directed therapy to deliver clinical services of excellence and evidence-based care to patients with cancer in the China GBA. METHODS Thirty experts used a modified Delphi method. The evidence extracted to support the statements was graded according to the GRADE system and reported according to the Revised Standards for Quality Improvement Reporting Excellence guidelines, version 2.0. RESULTS The POWG reached consensus in six key statements: harmonization of reporting and quality assurance of NGS; molecular tumor board and clinical decision support systems for PO; education and training; research and real-world data collection, patient engagement, regulations, and financial reimbursement of PO treatment strategies; and clinical recommendations and implementation of PO in clinical practice. CONCLUSION POWG consensus statements standardize the clinical application of NGS CGPs, streamline the interpretation of clinically significant genomic alterations, and align actionable mutations with sequence-directed therapies. The POWG consensus statements may harmonize the utility and delivery of PO in China's GBA.
Collapse
Affiliation(s)
- Tai-Chung Lam
- Department of Clinical Oncology, Queen Mary Hospital/Hong Kong University-Shenzhen Hospital, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | | | - Joseph Siu-Kie Au
- Adventist Oncology Centre, Hong Kong Adventist Hospital, Hong Kong SAR, China
| | - Edmond Shiu-Kwan Ma
- Clinical and Molecular Pathology and Cancer Genetics Centre, Hong Kong Sanatorium & Hospital, Hong Kong SAR, China
| | - Stephen Tak-Sum Lam
- Clinical Genetic Service Centre, Hong Kong Sanatorium & Hospital, Hong Kong SAR, China
| | - Herbert Ho-Fung Loong
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jason Wing Hon Wong
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - S.N. Michael Wong
- Department of Clinical Oncology, Queen Mary Hospital/Hong Kong University-Shenzhen Hospital, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Victor Ho-Fun Lee
- Department of Clinical Oncology, Queen Mary Hospital/Hong Kong University-Shenzhen Hospital, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | | | | | - Michael Tsz-Yeung Kam
- Department of Clinical Oncology, Pamela Youde Nethersole Eastern Hospital, Hong Kong SAR, China
| | | | - Fiona Mei-Ying Lim
- Department of Clinical Oncology, Princess Margaret Hospital, Hong Kong SAR, China
| | | | | | - Ka-Man Cheung
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | | | | | - Foon-Yiu Cheung
- Hong Kong International Oncology Centre, Hong Kong SAR, China
| | - Darren M.C. Poon
- Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, Hong Kong SAR, China
| | | | | | - Lawrence Wing-Chi Chan
- Department of Health Technology & Informatics, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Sze-Chuen Cesar Wong
- Department of Health Technology & Informatics, Hong Kong Polytechnic University, Hong Kong SAR, China
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Ya-Bing Cao
- Department of Radiology & Oncology, Kiang Wu Hospital, Macao SAR, China
| | - Cheng-Vai Hui
- Department of Clinical Oncology, Centro Hospitalar Conde de São Januário, Macao SAR, China
| | - Jack Zhi-Jian Chen
- Department of Radiation Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Shenzhen Center, Shenzhen, China
| | - Jian-Hua Chang
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Shenzhen Center, Shenzhen, China
| | - Spring Feng-Ming Kong
- Department of Clinical Oncology, Queen Mary Hospital/Hong Kong University-Shenzhen Hospital, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Aya El Helali
- Department of Clinical Oncology, Queen Mary Hospital/Hong Kong University-Shenzhen Hospital, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
4
|
Abstract
Mitochondrial diseases require customized approaches for reproductive counseling, addressing differences in recurrence risks and reproductive options. The majority of mitochondrial diseases is caused by mutations in nuclear genes and segregate in a Mendelian way. Prenatal diagnosis (PND) or preimplantation genetic testing (PGT) are available to prevent the birth of another severely affected child. In at least 15%-25% of cases, mitochondrial diseases are caused by mitochondrial DNA (mtDNA) mutations, which can occur de novo (25%) or be maternally inherited. For de novo mtDNA mutations, the recurrence risk is low and PND can be offered for reassurance. For maternally inherited, heteroplasmic mtDNA mutations, the recurrence risk is often unpredictable, due to the mitochondrial bottleneck. PND for mtDNA mutations is technically possible, but often not applicable given limitations in predicting the phenotype. Another option for preventing the transmission of mtDNA diseases is PGT. Embryos with mutant load below the expression threshold are being transferred. Oocyte donation is another safe option to prevent the transmission of mtDNA disease to a future child for couples who reject PGT. Recently, mitochondrial replacement therapy (MRT) became available for clinical application as an alternative to prevent the transmission of heteroplasmic and homoplasmic mtDNA mutations.
Collapse
|
5
|
Identification of Germline Variants in Patients with Hereditary Cancer Syndromes in Northeast Mexico. Genes (Basel) 2023; 14:genes14020341. [PMID: 36833268 PMCID: PMC9957276 DOI: 10.3390/genes14020341] [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: 11/18/2022] [Revised: 01/12/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Hereditary cancer syndromes (HCS) are genetic diseases with an increased risk of developing cancer. This research describes the implementation of a cancer prevention model, genetic counseling, and germline variants testing in an oncologic center in Mexico. A total of 315 patients received genetic counseling, genetic testing was offered, and 205 individuals were tested for HCS. In 6 years, 131 (63.90%) probands and 74 (36.09%) relatives were tested. Among the probands, we found that 85 (63.9%) had at least one germline variant. We identified founder mutations in BRCA1 and a novel variant in APC that led to the creation of an in-house detection process for the whole family. The most frequent syndrome was hereditary breast and ovarian cancer syndrome (HBOC) (41 cases with BRCA1 germline variants in most of the cases), followed by eight cases of hereditary non-polyposic cancer syndrome (HNPCC or Lynch syndrome) (with MLH1 as the primarily responsible gene), and other high cancer risk syndromes. Genetic counseling in HCS is still a global challenge. Multigene panels are an essential tool to detect the variants frequency. Our program has a high detection rate of probands with HCS and pathogenic variants (40%), compared with other reports that detect 10% in other populations.
Collapse
|
6
|
Bon SBB, Wouters RHP, Hol JA, Jongmans MCJ, van den Heuvel‐Eibrink MM, Grootenhuis MA. Parents' experiences with large-scale sequencing for genetic predisposition in pediatric renal cancer: A qualitative study. Psychooncology 2022; 31:1692-1699. [PMID: 35962481 PMCID: PMC9804506 DOI: 10.1002/pon.6016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/12/2022] [Accepted: 08/07/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVE In pediatric oncology, large-scale genetic sequencing contributes to the identification of cancer predisposition, which can facilitate surveillance and family counseling. Our qualitative study explores families' motives, knowledge, and views regarding germline genetic sequencing to improve future counseling and support. METHODS Semi-structured interviews were conducted with parents of children with renal tumors participating in a national center, germline sequencing study. An inductive thematic analysis approach was used. Twenty nine parents participated, 17 mothers and 12 fathers. The median age of the affected children was 4 years. RESULTS Parents were generally positive about sequencing and reported a combination of individual and altruistic motives to participate. Some families counseled about sequencing shortly after cancer diagnosis felt overwhelmed. Many parents had difficulties distinguishing between panel and exome-wide analysis. Families in which no predisposition was identified felt reassured. Most families did not experience distress after a predisposition was disclosed, although sometimes stress following disclosure of a predisposition added to pre-existing (cancer-related) stress. CONCLUSIONS Even though families reported positive experiences with germline genetic sequencing to detect cancer predisposition, timing of consent for sequencing as well as parents' understanding of genetic concepts can be further improved.
Collapse
Affiliation(s)
| | | | - Janna A. Hol
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | - Marjolijn C. J. Jongmans
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Department of GeneticsUniversity Medical Center UtrechtUtrechtThe Netherlands,Division of Child HealthUMCU‐Wilhelmina's Children's HospitalUtrechtThe Netherlands
| | - Marry M. van den Heuvel‐Eibrink
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Division of Child HealthUMCU‐Wilhelmina's Children's HospitalUtrechtThe Netherlands
| | - Martha A. Grootenhuis
- Princess Máxima Center for Pediatric OncologyUtrechtThe Netherlands,Division of Child HealthUMCU‐Wilhelmina's Children's HospitalUtrechtThe Netherlands
| |
Collapse
|
7
|
Luthra A, Mastrogiacomo B, Smith SA, Chakravarty D, Schultz N, Sanchez-Vega F. Computational methods and translational applications for targeted next-generation sequencing platforms. Genes Chromosomes Cancer 2022; 61:322-331. [PMID: 35066956 PMCID: PMC10129038 DOI: 10.1002/gcc.23023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 01/10/2022] [Indexed: 11/09/2022] Open
Abstract
During the past decade, next-generation sequencing (NGS) technologies have become widely adopted in cancer research and clinical care. Common applications within the clinical setting include patient stratification into relevant molecular subtypes, identification of biomarkers of response and resistance to targeted and systemic therapies, assessment of heritable cancer risk based on known pathogenic variants, and longitudinal monitoring of treatment response. The need for efficient downstream processing and reliable interpretation of sequencing data has led to the development of novel algorithms and computational pipelines, as well as structured knowledge bases that link genomic alterations to currently available drugs and ongoing clinical trials. Cancer centers around the world use different types of targeted solid-tissue and blood based NGS assays to analyze the genomic and transcriptomic profile of patients as part of their routine clinical care. Recently, cross-institutional collaborations have led to the creation of large pooled datasets that can offer valuable insights into the genomics of rare cancers.
Collapse
Affiliation(s)
- Anisha Luthra
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Brooke Mastrogiacomo
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Shaleigh A Smith
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Debyani Chakravarty
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Nikolaus Schultz
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Francisco Sanchez-Vega
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
8
|
Kisiangani I, Mohamed SF, Kyobutungi C, Tindana P, Ghansah A, Ramsay M, Asiki G. Perspectives on returning individual and aggregate genomic research results to study participants and communities in Kenya: a qualitative study. BMC Med Ethics 2022; 23:27. [PMID: 35300680 PMCID: PMC8932129 DOI: 10.1186/s12910-022-00767-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 03/08/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A fundamental ethical challenge in conducting genomics research is the question of what and how individual level genetic findings and aggregate genomic results should be conveyed to research participants and communities. This is within the context of minimal guidance, policies, and experiences, particularly in Africa. The aim of this study was to explore the perspectives of key stakeholders' on returning genomics research results to participants in Kenya. METHODS This qualitative study involved focus group discussions (FGDs) and in-depth interviews (IDIs) with 69 stakeholders. The purposively selected participants, included research ethics committee (REC) members (8), community members (44), community resource persons (8), and researchers (9). A semi-structured interview guide was used to facilitate discussions. Six FGDs and twenty-five (IDIs) were conducted among the different stakeholders. The issues explored in the interviews included: (1) views on returning results, (2) kind of results to be returned, (3) value of returning results to participants, and (4) challenges anticipated in returning results to participants and communities. The interviews were audio-recorded, transcribed verbatim, and coded in Nvivo 12 pro. Thematic and content analysis was conducted. RESULTS Participants agreed on the importance of returning genomic results either as individual or aggregate results. The most cited reasons for returning of genomic results included recognizing participants' contribution to research, encouraging participation in future research, and increasing the awareness of scientific progress. Other aspects on how genomic research results should be shared included sharing easy to understand results in the shortest time possible and maintaining confidentiality when sharing sensitive results. CONCLUSIONS This study identified key stakeholders' perspectives on returning genomic results at the individual and community levels in two urban informal settlements of Nairobi. The majority of the participants expect to receive feedback about their genomic results, and it is an obligation for researchers to see how to best fulfil it.
Collapse
Affiliation(s)
- Isaac Kisiangani
- African Population and Health Research Center (APHRC), P.O. Box 10787, Nairobi, 00100 Kenya
| | - Shukri F. Mohamed
- African Population and Health Research Center (APHRC), P.O. Box 10787, Nairobi, 00100 Kenya
| | - Catherine Kyobutungi
- African Population and Health Research Center (APHRC), P.O. Box 10787, Nairobi, 00100 Kenya
| | - Paulina Tindana
- School of Public Health, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Anita Ghansah
- Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Ghana
| | - Michele Ramsay
- Sydney Brenner Institute for Molecular Bioscience, The University of Witwatersrand, The Mount, 9 Jubilee Rd, Parktown, Johannesburg, 2193 South Africa
| | - Gershim Asiki
- African Population and Health Research Center (APHRC), P.O. Box 10787, Nairobi, 00100 Kenya
| |
Collapse
|
9
|
Ochieng J, Kwagala B, Barugahare J, Mwaka E, Ekusai-Sebatta D, Ali J, Sewankambo NK. Perspectives and ethical considerations for return of genetics and genomics research results: a qualitative study of genomics researchers in Uganda. BMC Med Ethics 2021; 22:154. [PMID: 34798900 PMCID: PMC8603565 DOI: 10.1186/s12910-021-00724-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 11/03/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The return of genetics and genomics research results has been a subject of ongoing global debate. Such feedback is ethically desirable to update participants on research findings particularly those deemed clinically significant. Although there is limited literature, debate continues in African on what constitutes appropriate practice regarding the return of results for genetics and genomics research. This study explored perspectives and ethical considerations of Ugandan genomics researchers regarding the return of genetics and genomics research results. METHODS This was a qualitative study that employed in-depth interviews. Thirty participants were purposively selected based on their expertise as genomics researchers in Uganda. Data were analysed through content analysis along the main themes of the study using a comprehensive thematic matrix, to identify common patterns arising from the narratives. NVivo software 12 was used to support data analysis. RESULTS The return of genetics and genomics research results was generally acceptable to researchers, and some indicated that they had previously returned individual or aggregate results to participants and communities. The main reasons cited for sharing research results with participants included their clinical utility, actionability and overall benefit to society. Ethical considerations for appropriate return of results included a need for effective community engagement, genetic counselling prior to disclosure of the results, adequate informed consent, and proper assessment of the implications of, or consequences of returning of results. However, the approaches to return of results were perceived as unstandardized due to the lack of appropriate regulatory frameworks. CONCLUSIONS The return of genetic and genomic research results is generally acceptable to researchers despite the lack of appropriate regulatory frameworks. Ethical considerations for return of genetics and genomics research results are highly divergent, hence the need for national ethical guidelines to appropriately regulate the practice.
Collapse
Affiliation(s)
- Joseph Ochieng
- Department of Anatomy, College of Health Sciences, School of Biomedical Sciences, Makerere University, P.O Box 7072, Kampala, Uganda.
| | - Betty Kwagala
- School of Business and Management Studies, Makerere University, Kampala, Uganda
| | | | - Erisa Mwaka
- Department of Anatomy, College of Health Sciences, School of Biomedical Sciences, Makerere University, P.O Box 7072, Kampala, Uganda
| | - Deborah Ekusai-Sebatta
- Department of Anatomy, College of Health Sciences, School of Biomedical Sciences, Makerere University, P.O Box 7072, Kampala, Uganda
| | - Joseph Ali
- Johns Hopkins Bloomberg School of Public Health, Baltimore, USA
- Johns Hopkins Berman Institute of Bioethics, Baltimore, USA
| | | |
Collapse
|
10
|
Abstract
Technological innovation and rapid reduction in sequencing costs have enabled the genomic profiling of hundreds of cancer-associated genes as a component of routine cancer care. Tumour genomic profiling can refine cancer subtype classification, identify which patients are most likely to benefit from systemic therapies and screen for germline variants that influence heritable cancer risk. Here, we discuss ongoing efforts to enhance the clinical utility of tumour genomic profiling by integrating tumour and germline analyses, characterizing allelic context and identifying mutational signatures that influence therapy response. We also discuss the potential clinical utility of more comprehensive whole-genome and whole-transcriptome sequencing and ultra-sensitive cell-free DNA profiling platforms, which allow for minimally invasive, serial analyses of tumour-derived DNA in blood.
Collapse
Affiliation(s)
- Debyani Chakravarty
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David B Solit
- Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA. .,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
11
|
Bijlsma R, Wouters R, Wessels H, Sleijfer S, Beerepoot L, Ten Bokkel Huinink D, Cruijsen H, Heijns J, Lolkema MP, Steeghs N, van Voorthuizen T, Vulink A, Witteveen E, Ausems M, Bredenoord A, May AM, Voest E. Preferences to receive unsolicited findings of germline genome sequencing in a large population of patients with cancer. ESMO Open 2021; 5:S2059-7029(20)30053-3. [PMID: 32312756 PMCID: PMC7200077 DOI: 10.1136/esmoopen-2019-000619] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/05/2019] [Accepted: 12/29/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND In precision medicine, somatic and germline DNA sequencing are essential to make genome-guided treatment decisions in patients with cancer. However, it can also uncover unsolicited findings (UFs) in germline DNA that could have a substantial impact on the lives of patients and their relatives. It is therefore critical to understand the preferences of patients with cancer concerning UFs derived from whole-exome (WES) or whole-genome sequencing (WGS). METHODS In a quantitative multicentre study, adult patients with cancer (any stage and origin of disease) were surveyed through a digital questionnaire based on previous semi-structured interviews. Background knowledge was provided by showing two videos, introducing basic concepts of genetics and general information about different categories of UFs (actionable, non-actionable, reproductive significance, unknown significance). RESULTS In total 1072 patients were included of whom 701 participants completed the whole questionnaire. Overall, 686 (85.1%) participants wanted to be informed about UFs in general. After introduction of four UFs categories, 113 participants (14.8%) changed their answer: 718 (94.2%) participants opted for actionable variants, 537 (72.4%) for non-actionable variants, 635 (87.0%) participants for UFs of reproductive significance and 521 (71.8%) for UFs of unknown significance. Men were more interested in receiving certain UFs than women: non-actionable: OR 3.32; 95% CI 2.05 to 5.37, reproductive significance: OR 1.97; 95% CI 1.05 to 3.67 and unknown significance: OR 2.00; 95% CI 1.25 to 3.21. In total, 244 (33%) participants conceded family members to have access to their UFs while still alive. 603 (82%) participants agreed to information being shared with relatives, after they would pass away. CONCLUSION Our study showed that the vast majority of patients with cancer desires to receive all UFs of genome testing, although a substantial minority does not wish to receive non-actionable findings. Incorporation of categories in informed consent procedures supports patients in making informed decisions on UFs.
Collapse
Affiliation(s)
- Rhode Bijlsma
- Department of Medical Oncology, University Medical Center Utrecht, Cancer Center, Utrecht, The Netherlands
| | - Roel Wouters
- Department of Medical Humanities, University Medical Center Utrecht, Julius Center, Utrecht, The Netherlands
| | - Hester Wessels
- Department of Corporate Communications, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.,Center for Personalized Cancer Treatment (CPCT), Rotterdam, The Netherlands
| | - Laurens Beerepoot
- Department of Medical Oncology, Elisabeth-Tweesteden Hospital, Tilburg, The Netherlands
| | | | - Hester Cruijsen
- Department of Medical Oncology, Antonius Hospital, Sneek, The Netherlands
| | - Joan Heijns
- Department of Medical Oncology, Amphia Hospital, Breda, The Netherlands
| | - Martijn P Lolkema
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Annelie Vulink
- Department of Medical Oncology, Reinier de Graaf Gasthuis, Delft, The Netherlands
| | - Els Witteveen
- Department of Medical Oncology, University Medical Center Utrecht, Cancer Center, Utrecht, The Netherlands
| | - Margreet Ausems
- Department of Genetics, Division Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Annelien Bredenoord
- Department of Medical Humanities, University Medical Center Utrecht, Julius Center, Utrecht, The Netherlands
| | - Anne M May
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Emile Voest
- Center for Personalized Cancer Treatment (CPCT), Rotterdam, The Netherlands .,Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Roosan D, Hwang A, Roosan MR. Pharmacogenomics cascade testing (PhaCT): a novel approach for preemptive pharmacogenomics testing to optimize medication therapy. THE PHARMACOGENOMICS JOURNAL 2021; 21:1-7. [PMID: 32843688 PMCID: PMC7840503 DOI: 10.1038/s41397-020-00182-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 06/18/2020] [Accepted: 08/12/2020] [Indexed: 11/08/2022]
Abstract
The implementation of pharmacogenomics (PGx) has come a long way since the dawn of utilizing pharmacogenomic data in clinical patient care. However, the potential benefits of sharing PGx results have yet to be explored. In this paper, we explore the willingness of patients to share PGx results, as well as the inclusion of family medication history in identifying potential family members for pharmacogenomics cascade testing (PhaCT). The genetic similarities in families allow for identifying potential gene variants prior to official preemptive testing. Once a candidate patient is determined, PhaCT can be initiated. PhaCT recognizes that further cascade testing throughout a family can serve to improve precision medicine. In order to make PhaCT feasible, we propose a novel shareable HIPAA-compliant informatics platform that will enable patients to manage not only their own test results and medications but also those of their family members. The informatics platform will be an external genomics system with capabilities to integrate with patients' electronic health records. Patients will be given the tools to provide information to and work with clinicians in identifying family members for PhaCT through this platform. Offering patients the tools to share PGx results with their family members for preemptive testing could be the key to empowering patients. Clinicians can utilize PhaCT to potentially improve medication adherence, which may consequently help to distribute the burden of health management between patients, family members, providers, and payers.
Collapse
Affiliation(s)
- Don Roosan
- Department of Pharmacy Practice and Administration, College of Pharmacy, Western University of Health Sciences, Pomona, CA, USA.
| | - Angela Hwang
- Department of Pharmacy Practice and Administration, College of Pharmacy, Western University of Health Sciences, Pomona, CA, USA
| | - Moom R Roosan
- Department of Pharmacy Practice, School of Pharmacy, Chapman University, School of Pharmacy, Irvine, CA, USA.
| |
Collapse
|
13
|
Bianco B, Barbosa CP, Trevisan CM, Laganà AS, Montagna E. Endometrial cancer: a genetic point of view. Transl Cancer Res 2020; 9:7706-7715. [PMID: 35117373 PMCID: PMC8797944 DOI: 10.21037/tcr-20-2334] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Endometrial cancer is the fourth most frequent gynecological cancer and the most frequent type of uterine cancer. There is an increase in the incidence and mortality of uterine cancers in the past few decades, and there are no well-established screening programs for endometrial cancer currently. Most endometrial cancers arise through the interplay of familial, genetic, and lifestyle factors. Although a number of genetic factors modify endometrial cancer susceptibility, they are not of standard use in the clinical assessment of prognosis. We conducted a comprehensive systematic literature review to provide an overview of the relationship between genetic factors and risk for endometrial cancer. METHODS MEDLINE and EMBASE databases were searched for studies between January 2010 to March 2020 reporting the genes associated with endometrial cancer. RESULTS Through the selection process, we retrieved 186 studies comprising 329 genes identified using several molecular methodologies in all human chromosomes and in mitochondrial DNA. Endometrial cancer exhibits a molecular complexity and heterogeneity coherent with its clinical and histologic variability. Improved characterization of molecular alterations of each histological type provides relevant information about the prognosis and potential response to new therapies. CONCLUSIONS The current challenge is the integration of clinicopathologic and molecular factors to improve the diagnosis, prognosis, and treatment of endometrial cancer.
Collapse
Affiliation(s)
- Bianca Bianco
- Discipline of Sexual and Reproductive Health and Populational Genetics, Department of Collective Health, Santo André, Brazil
| | - Caio Parente Barbosa
- Discipline of Sexual and Reproductive Health and Populational Genetics, Department of Collective Health, Santo André, Brazil
| | | | - Antonio Simone Laganà
- Department of Obstetrics and Gynecology, “Filippo Del Ponte” Hospital, University of Insubria, Varese, Italy
| | - Erik Montagna
- Postgraduate Program in Health Sciences, Faculdade de Medicina do ABC, Santo André, Brazil
| |
Collapse
|
14
|
Ethical challenges of precision cancer medicine. Semin Cancer Biol 2020; 84:263-270. [PMID: 33045356 DOI: 10.1016/j.semcancer.2020.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 08/25/2020] [Accepted: 09/20/2020] [Indexed: 11/21/2022]
Abstract
Amongst common diseases, cancer is often both a leader in self-regulatory policy, or the field for contentious ethical issues such as the patenting of the BRCA1/2 genes. With the advent of genomic sequencing technologies, achieving precision cancer medicine requires prospective norms due to the large and varied sources of data involved. Here, we discuss the ethical and legal aspects of the policy debate around the relevant topics in precision cancer medicine: the return of incidental findings and sequencing raw data to patients, the communication of genetic results to patients' relatives, privacy and communication risks with concomitant oversight strategies, patient participation and consent models. We present the arguments and empirical data supporting specific policy solutions delineating still contested areas. What type of consent and oversight are required to acquire genomic data or to access it where desired, either by the participant/patient or third-party researchers? Most of the raw sequencing data is still uninterpretable and the variants revealed subject to reinterpretation over time. No doubt the ethical challenges of precision cancer medicine are a prototype of what's to come for other diseases. They are also paradigmatic for regulatory and ethical questions of the translational endeavors since the two worlds - basic science and patient care - are governed by different ethical and legal principles that need to be reconciled in precision cancer medicine.
Collapse
|
15
|
Smetana GW, Vassy JL, Hofstatter E, Libman H. Should You Recommend Direct-to-Consumer Genetic Testing for This Patient? : Grand Rounds Discussion From Beth Israel Deaconess Medical Center. Ann Intern Med 2020; 173:563-571. [PMID: 33017547 DOI: 10.7326/m20-5419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In recent years, the number of patients choosing to have direct-to-consumer (DTC) genetic testing without involving their clinicians has increased substantially. For example, the number of subscribers to a commonly used testing site has grown to more than 10 million. These services have been heavily marketed in the United States and often include information about ancestry; genetic traits; and, increasingly, disease risk. In clinical care, genetic testing by a physician is accompanied by both pre- and posttest counseling by a trained genetic counselor. However, there are not enough genetic counselors to meet the needs of all persons contemplating DTC genetic testing. Formal genetic counseling includes preparation of a family pedigree; a discussion about potential benefits, the possibility that some information might be stressful to receive or difficult to understand, and the potential for disclosure of genetic information; and a detailed informed consent process. Some DTC tests for genetic susceptibilities look for only a few known mutations in a particular gene (such as BRCA1); a negative test result does not exclude the possibility of a clinically important mutation. A positive DTC genetic test result that might change clinical management should be followed by a confirmatory test through a genetics laboratory. Here, 2 expert physicians-a general internist and a medical oncologist with genetics experience-discuss an approach to counseling a patient who is considering DTC testing to learn more about his ancestry and his risk for metabolic syndrome.
Collapse
Affiliation(s)
- Gerald W Smetana
- Beth Israel Deaconess Medical Center, Boston, Massachusetts (G.W.S., H.L.)
| | - Jason L Vassy
- Harvard Medical School at Veterans Affairs Boston Healthcare System, Boston, Massachusetts (J.L.V.)
| | | | - Howard Libman
- Beth Israel Deaconess Medical Center, Boston, Massachusetts (G.W.S., H.L.)
| |
Collapse
|
16
|
Torrorey-Sawe R, van der Merwe N, Mining SK, Kotze MJ. Pioneering Informed Consent for Return of Research Results to Breast Cancer Patients Facing Barriers to Implementation of Genomic Medicine: The Kenyan BRCA1/2 Testing Experience Using Whole Exome Sequencing. Front Genet 2020; 11:170. [PMID: 32231682 PMCID: PMC7089032 DOI: 10.3389/fgene.2020.00170] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/12/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Obtaining informed consent from study participants and disseminating the findings responsibly is a key principle required for ethically conducted clinical and genetic research. Reports from African researchers providing feedback on insights gained during the return of whole exome sequencing (WES) results to breast cancer patients treated in resource-limited settings is lacking. AIM The empirical process used to fill this gap in relation to BRCA1/2 variant detection using WES provided unique insights incorporated into a pathology-supported genetic testing algorithm for return of research results to Kenyan breast cancer patients. METHODS The Informed consent form approved by the Moi Teaching and Referral Hospital in Kenya was adopted from a translational research study conducted in South Africa. Initially, the informed consent process was piloted in 16 Kenyan female patients referred for breast surgery, following a community-based awareness campaign. A total of 95 female and two male breast cancer patients were enrolled in the study from 2013 to 2016. Immunohistochemistry (IHC) results of estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor-2 (HER2) status were obtained from hospital records. DNA of patients with a family history of cancer was extracted from saliva and screened for pathogenic variants in the BRCA1/2 genes as the first step using WES. RESULTS Ten patients approached for participation in this study declined to sign the informed consent form. Data on IHC used as a proxy for molecular subtype were available in 8 of 13 breast cancer patients (62%) with a family history of cancer. Five BRCA1/2 variants of uncertain clinical significance were detected, as well as a pathogenic BRCA2 variant (c.5159C > A; S1720∗) in a female patient eligible for return of WES results. CONCLUSION Experience gained during the qualitative pilot phase was essential to overcome challenges associated with the translation of sophisticated genetic terms into native African languages. Detection of a pathogenic BRCA2 variant in a patient with familial breast cancer, frequently associated with hormone receptor-positive breast carcinoma as reported in this case, led to a high level of confidence on which to base risk management in future. Implementation of new technologies alongside standard pathology provides a practical approach to the application of genomic medicine in Africa.
Collapse
Affiliation(s)
- Rispah Torrorey-Sawe
- Division of Chemical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
- Department of Immunology, School of Medicine, College of Health Sciences, Moi University, Eldoret, Kenya
| | - Nicole van der Merwe
- Division of Chemical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Simeon Kipkoech Mining
- Department of Immunology, School of Medicine, College of Health Sciences, Moi University, Eldoret, Kenya
| | - Maritha J Kotze
- Division of Chemical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
- National Health Laboratory Service, Tygerberg Hospital, Cape Town, South Africa
| |
Collapse
|
17
|
Ozcan A, Erdogan S, Truong LD. Hereditary Syndromes Associated with Kidney Tumors. KIDNEY CANCER 2020. [DOI: 10.1007/978-3-030-28333-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
18
|
Kimberly RP. Translational Research-For the Individual and the Community. J Health Care Poor Underserved 2019; 30:79-85. [PMID: 31735721 DOI: 10.1353/hpu.2019.0118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The potential for translational research to improve human health is unprecedented, as the integration of genetic health risks with other data influencing health provides substantial opportunities for improvement. However, how integrating these data sources in a fair, unbiased and appropriate way without reinforcing pre-existing assumptions requires thoughtful implementation. Furthermore, integration of new technologies requires assessment of needs and benefits for the individual balanced with community needs and goals. Thus, examination of values, goals and implicit assumptions through transparent, authentic engagement of individuals and communities is essential.
Collapse
|
19
|
Zimmermann B, Elger B, Shaw D. Media Coverage of Ethical Issues in Predictive Genetic Testing: A Qualitative Analysis. AJOB Empir Bioeth 2019; 10:250-264. [PMID: 31596686 DOI: 10.1080/23294515.2019.1670275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background: Predictive genetic testing (PGT) raises many ethical issues and is of increasing interest to the general population. Mass media, especially newspapers, are the public's main source of information on this topic. Methods: We conducted a content analysis of British newspaper reporting, assessing which ethical issues were mentioned. The analysis was qualitative with semi-quantitative aspects. All articles about PGT published in The Guardian and the Daily Telegraph from 2011 to 2016 were included. Results: Most ethical issues discussed in the scientific and ethical literature are implicitly or explicitly covered in newspapers, but there was no discussion of incidental findings and the possibility of false reassurance of a negative test result was mentioned only once. There are also important gaps regarding the multidimensional nature and complexity of many issues. The Guardian mentioned ethical issues more frequently than the Daily Telegraph. Most ethical issues were portrayed as first-person narratives. Conclusions: Ethical issues concern potential test users and society more than scientific background knowledge about such tests; therefore, more efforts should be taken to address these complex issues in a manner that is comprehensible for the lay public.
Collapse
Affiliation(s)
- Bettina Zimmermann
- Institute for Biomedical Ethics, University of Basel , Basel , Switzerland
| | - Bernice Elger
- Institute for Biomedical Ethics, University of Basel , Basel , Switzerland
| | - David Shaw
- Institute for Biomedical Ethics, University of Basel , Basel , Switzerland
- Care and Public Health Research Institute, Maastricht University , Maastricht , the Netherlands
| |
Collapse
|
20
|
Kim JY, Byeon JS. Genetic Counseling and Surveillance Focused on Lynch Syndrome. JOURNAL OF THE ANUS RECTUM AND COLON 2019; 3:60-68. [PMID: 31559369 PMCID: PMC6752118 DOI: 10.23922/jarc.2019-002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 02/08/2019] [Indexed: 12/20/2022]
Abstract
Lynch syndrome is a hereditary cancer syndrome caused by germline mutations in one of several DNA mismatch repair genes. Lynch syndrome leads to an increased lifetime risk of various cancers, particularly colorectal, and endometrial cancers. After identifying patients suspected of having Lynch syndrome by clinical criteria, computational prediction models, and/or universal tumor testing, genetic testing is performed to confirm the diagnosis. Before and after genetic testing, genetic counseling should be provided. Genetic counseling should involve a detailed personal and family history, information on the disorder and genetic tests, discussion of the management and surveillance of the disease, career plan, family plan, and psychosocial support. Surveillance of colorectal cancer and other malignancies is of paramount importance for properly managing Lynch syndrome. This review focuses on important considerations in genetic counseling and the latest insights into the surveillance of individuals and families with Lynch syndrome.
Collapse
Affiliation(s)
- Jin Yong Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jeong-Sik Byeon
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
21
|
Bijlsma RM, Wessels H, Wouters RHP, May AM, Ausems MGEM, Voest EE, Bredenoord AL. Cancer patients' intentions towards receiving unsolicited genetic information obtained using next-generation sequencing. Fam Cancer 2019; 17:309-316. [PMID: 28852913 PMCID: PMC5893692 DOI: 10.1007/s10689-017-0033-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Next-generation sequencing (NGS) can be used to generate information about a patient’s tumour and personal genome. This powerful diagnostic tool provides solicited and unsolicited hereditary genetic (risk) information that could have consequences for cancer patients and their quality of life. A well-defined approach for returning appropriate genetic risk information is needed in personalized cancer care. A qualitative design with semi-structured interviews was used. We conducted interviews with 24 Dutch patients with different types of cancer, both NGS-experienced and NGS-inexperienced, to learn their intentions, needs and preferences towards receiving unsolicited genetic information obtained using NGS. Almost all participants had a positive attitude towards receiving unsolicited findings. After receiving comprehensive background information on NGS, including a binning model of four categories of unsolicited findings, most participants preferred to receive only subsets of genetic information. Their main concern was their own and others’ (including family members) ability to cope with (the increased risk of having) a genetic disorder. Providing background information gave cancer patients the opportunity to select subsets of findings and increased their ability to make an informed choice. Special attention is needed for social and emotional factors to support the patients themselves and when communicating test results with their family members.
Collapse
Affiliation(s)
- Rhodé M Bijlsma
- Department of Medical Oncology, Cancer Center, University Medical Center Utrecht, Q05.4.300, PO Box 85500, 3508 GA, Utrecht, The Netherlands.
| | - Hester Wessels
- Department of Corporate Communications, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roel H P Wouters
- Department of Medical Humanities, Julius Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Anne M May
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Emile E Voest
- Department of Medical Oncology, Netherlands Cancer Institute Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Annelien L Bredenoord
- Department of Medical Humanities, Julius Center, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
22
|
Abstract
OBJECTIVE To review ethical, legal, and social implications of genomics, a ground-breaking science that when applied improves cancer care outcomes. DATA SOURCES PubMed, Cumulative Index to Nursing and Allied Health (CINAHL), Cochrane Library, consensus statements, and professional guidelines. CONCLUSION Ethical, legal, and social domains of genomics are not fully delineated. Areas needing further discussion and policies include return of findings, informed consent, electronic health records, and data resources and sharing. IMPLICATIONS FOR NURSING PRACTICE All nurses need a basic understanding of the ethical, legal, and social implications of genomics.
Collapse
|
23
|
Atutornu J, Hayre CM. Personalised Medicine and Medical Imaging: Opportunities and Challenges for Contemporary Health Care. J Med Imaging Radiat Sci 2018; 49:352-359. [DOI: 10.1016/j.jmir.2018.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/04/2018] [Accepted: 07/04/2018] [Indexed: 01/09/2023]
|
24
|
Vos S, van Diest PJ, Ausems MGEM, van Dijk MR, de Leng WWJ, Bredenoord AL. Ethical considerations for modern molecular pathology. J Pathol 2018; 246:405-414. [DOI: 10.1002/path.5157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/03/2018] [Accepted: 08/14/2018] [Indexed: 01/08/2023]
Affiliation(s)
- Shoko Vos
- Department of Pathology; University Medical Center Utrecht; Utrecht The Netherlands
| | - Paul J van Diest
- Department of Pathology; University Medical Center Utrecht; Utrecht The Netherlands
| | - Margreet GEM Ausems
- Department of Medical Genetics; University Medical Center Utrecht; Utrecht The Netherlands
| | - Marijke R van Dijk
- Department of Pathology; University Medical Center Utrecht; Utrecht The Netherlands
| | - Wendy WJ de Leng
- Department of Pathology; University Medical Center Utrecht; Utrecht The Netherlands
| | - Annelien L Bredenoord
- Department of Medical Humanities; University Medical Center Utrecht; Utrecht The Netherlands
| |
Collapse
|
25
|
Rosier M, Guedj M, Calvas P, Julia S, Garnier C, Cambon-Thomsen A, Muñoz Sastre MT. French People’s Views on the Appropriateness of Disclosing an Unsolicited Finding in Medical Genetics: A Preliminary Study. UNIVERSITAS PSYCHOLOGICA 2018. [DOI: 10.11144/javeriana.upsy17-4.fpva] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
With progress in medical genetics, genome-sequencing techniques are becoming more and more efficient. However, these genetic tests may lead to the detection of unsolicited genetic findings, i.e. findings that are not the primary purpose of the screening. New ethical issues have emerged, in particular the question of whether to disclose these unsolicited findings to the patient or not. Forty-seven patients under supervision in a Medical Genetics service, 15 health professionals and 107 members of the French general population expressed their opinion regarding the appropriateness of disclosing an unsolicited high penetrance genetic finding in 36 scenarios containing three pieces of information on: a) patient information and consent;b) possibility of prevention and treatment of the detected genetic disease; and c) disclosure of the results by the physician (e.g., no disclosure of the unsolicited results). Four positions were found that were called Respect for patient’s autonomy, Beneficence to patient, Non-maleficence, and Always appropriate.
Collapse
|
26
|
Van Valckenborgh E, Hébrant A, Antoniou A, Van Hoof W, Van Bussel J, Pauwels P, Salgado R, Van Doren W, Waeytens A, Van den Bulcke M. Roadbook for the implementation of next-generation sequencing in clinical practice in oncology and hemato-oncology in Belgium. ACTA ACUST UNITED AC 2018; 76:49. [PMID: 30202522 PMCID: PMC6125873 DOI: 10.1186/s13690-018-0295-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 07/10/2018] [Indexed: 11/30/2022]
Abstract
In the field of oncology research, next-generation sequencing has contributed significantly to the discovery of DNA mutations associated with diagnosis and prognosis. It also aids in the development of targeted therapies to specific mutations and the rise of personalized medicine. As part of molecular diagnostics in cancer patients, analysis by next-generation sequencing is becoming part of routine clinical practice. The introduction of this complex technology in a healthcare system comes with multiple challenges and requires a clear action plan. Such an action plan, as outlined in this paper, was developed in Belgium and includes steps in ensuring the quality and indications of NGS testing, installing data registration and tackling ethical issues. A final step is to perform a pilot study to control the access, quality, harmonization and expertise in DNA testing. This action plan can serve as a guide for similar initiatives by other countries to facilitate NGS implementation in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | - Patrick Pauwels
- 4Pathology Department, University Hospital of Antwerp, Antwerp, Belgium
| | - Roberto Salgado
- Department of Pathology, GZA Hospitals Antwerp, Antwerp, Belgium.,Division of Research, Peter Mac Callum Cancer Center, Melbourne, Australia.,7Breast Cancer Translational Research Laboratory, Jules Bordet Institute, Brussels, Belgium
| | - Waltruda Van Doren
- Health Care Department, National Institute for Health and Disability Insurance, Brussels, Belgium
| | - Anouk Waeytens
- Health Care Department, National Institute for Health and Disability Insurance, Brussels, Belgium
| | | |
Collapse
|
27
|
van der Velden DL, van Herpen CML, van Laarhoven HWM, Smit EF, Groen HJM, Willems SM, Nederlof PM, Langenberg MHG, Cuppen E, Sleijfer S, Steeghs N, Voest EE. Molecular Tumor Boards: current practice and future needs. Ann Oncol 2018; 28:3070-3075. [PMID: 29045504 DOI: 10.1093/annonc/mdx528] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Due to rapid technical advances, steeply declining sequencing costs, and the ever-increasing number of targeted therapies, it can be expected that extensive tumor sequencing such as whole-exome and whole-genome sequencing will soon be applied in standard care. Clinicians will thus be confronted with increasingly complex genetic information and multiple test-platforms to choose from. General medical training, meanwhile, can hardly keep up with the pace of innovation. Consequently, there is a rapidly growing gap between clinical knowledge and genetic potential in cancer care. Multidisciplinary Molecular Tumor Boards (MTBs) have been suggested as a means to address this disparity, but shared experiences are scarce in literature and no quality requirements or guidelines have been published to date. Methods Based on literature review, a survey among hospitals in The Netherlands, and our own experience with the establishment of a nationally operating MTB, this article evaluates current knowledge and unmet needs and lays out a strategy for successful MTB implementation. Results Having access to an MTB can improve and increase the application of genetics-guided cancer care. In our survey, however, <50% of hospitals and only 5% of nonacademic hospitals had access to an MTB. In addition, current MTBs vary widely in terms of composition, tasks, tools, and workflow. This may not only lead to variation in quality of care but also hinders data sharing and thus creation of an effective learning community. Conclusions This article acknowledges a leading role for MTBs to govern (extensive) tumor sequencing into daily practice and proposes three basic necessities for successful MTB implementation: (i) global harmonization in cancer sequencing practices and procedures, (ii) minimal member and operational requirements, and (iii) an appropriate unsolicited findings policy. Meeting these prerequisites would not only optimize MTB functioning but also improve general interpretation and application of genomics-guided cancer care.
Collapse
Affiliation(s)
| | - C M L van Herpen
- Division of Medical Oncology, Radboud University Medical Center, Nijmegen
| | | | - E F Smit
- Division of Medical Oncology, Netherlands Cancer Institute, Amsterdam
| | - H J M Groen
- Division of Medical Oncology, University Medical Center Groningen, Groningen
| | - S M Willems
- Division of Pathology, University Medical Center Utrecht, Utrecht
| | - P M Nederlof
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam
| | | | - E Cuppen
- Human Genetics, University Medical Center Utrecht, Utrecht
| | - S Sleijfer
- Division of Medical Oncology, Erasmus University Medical Center, Rotterdam
| | - N Steeghs
- Division of Medical Oncology and Clinical Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - E E Voest
- Division of Molecular Oncology, Netherlands Cancer Institute, Amsterdam
| |
Collapse
|
28
|
Warner TD, Weil CJ, Andry C, Degenholtz HB, Parker L, Carithers LJ, Feige M, Wendler D, Pentz RD. Broad Consent for Research on Biospecimens: The Views of Actual Donors at Four U.S. Medical Centers. J Empir Res Hum Res Ethics 2018; 13:115-124. [PMID: 29390947 PMCID: PMC5869128 DOI: 10.1177/1556264617751204] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Commentators are concerned that broad consent may not provide biospecimen donors with sufficient information regarding possible future research uses of their tissue. We surveyed with interviews 302 cancer patients who had recently provided broad consent at four diverse academic medical centers. The majority of donors believed that the consent form provided them with sufficient information regarding future possible uses of their biospecimens. Donors expressed very positive views regarding tissue donation in general and endorsed the use of their biospecimens in future research across a wide range of contexts. Concerns regarding future uses were limited to for-profit research and research by investigators in other countries. These results support the use of broad consent to store and use biological samples in future research.
Collapse
Affiliation(s)
| | | | | | | | | | - Latarsha J. Carithers
- National Cancer Institute, National Institutes of Health (USA)
- Now at the National Institute of Dental & Craniofacial Research (USA)
| | - Michelle Feige
- Association for the Accreditation of Human Research Protection Programs, Inc. (USA)
| | | | - Rebecca D. Pentz
- Winship Cancer Institute, Emory University School of Medicine (USA)
| |
Collapse
|
29
|
Bijlsma RM, Wouters RHP, Wessels H, May AM, Ausems MGEM, Voest EE, Bredenoord AL. Managing unsolicited findings in genomics: A qualitative interview study with cancer patients. Psychooncology 2018; 27:1327-1333. [PMID: 29471587 DOI: 10.1002/pon.4676] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 02/08/2018] [Accepted: 02/10/2018] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Next-generation sequencing (NGS) is increasingly being employed in the context of personalized cancer treatment. Anticipating unsolicited findings that may arise during a NGS procedure is a key consideration; however, little is known about cancer patients' intentions, needs, and preferences concerning the return of unsolicited findings. METHODS A qualitative design using individual semi-structured interviews with 24 cancer patients was utilized to explore patients' decisions on whether to receive unsolicited findings from NGS. These interviews were subsequently analyzed using the constant comparative method to develop codes and themes. RESULTS We identified 4 interrelated themes that emerged in the context of the return of unsolicited findings. First, we describe how cancer patients expressed a strong need to control their lives. Second, we show the importance of family dynamics. Third, the NGS procedure regarding unsolicited findings is perceived as cognitively complex, and fourth, the procedure is also considered emotionally complex. CONCLUSIONS The results of our study contribute to a better understanding of what cancer patients consider important and what may motivate and influence them when making decisions on the disclosure of unsolicited findings following NGS. We show how Joel Feinberg's classification of autonomy may help clinicians to better understand cancer patients' desire for autonomous decision making while also acknowledging the emotional and cognitive difficulties regarding the disclosure of unsolicited findings. These insights could be helpful for clinicians to guide patients through this complex process.
Collapse
Affiliation(s)
- R M Bijlsma
- Cancer Center, Department of Medical Oncology, University Medical Center Utrecht, Utrecht, Netherlands
| | - R H P Wouters
- Julius Center, Department of Medical Humanities, University Medical Center Utrecht, Utrecht, Netherlands
| | - H Wessels
- Department of Corporate Communications, University Medical Center Utrecht, Utrecht, Netherlands
| | - A M May
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, Netherlands
| | - M G E M Ausems
- Department of Genetics, University Medical Center Utrecht, Utrecht, Netherlands
| | - E E Voest
- Netherlands Cancer Institute Antoni van Leeuwenhoek, Department of Medical Oncology, Amsterdam, Netherlands
| | - A L Bredenoord
- Julius Center, Department of Medical Humanities, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
30
|
Sendorek DH, Caloian C, Ellrott K, Bare JC, Yamaguchi TN, Ewing AD, Houlahan KE, Norman TC, Margolin AA, Stuart JM, Boutros PC. Germline contamination and leakage in whole genome somatic single nucleotide variant detection. BMC Bioinformatics 2018; 19:28. [PMID: 29385983 PMCID: PMC5793408 DOI: 10.1186/s12859-018-2046-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/24/2018] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The clinical sequencing of cancer genomes to personalize therapy is becoming routine across the world. However, concerns over patient re-identification from these data lead to questions about how tightly access should be controlled. It is not thought to be possible to re-identify patients from somatic variant data. However, somatic variant detection pipelines can mistakenly identify germline variants as somatic ones, a process called "germline leakage". The rate of germline leakage across different somatic variant detection pipelines is not well-understood, and it is uncertain whether or not somatic variant calls should be considered re-identifiable. To fill this gap, we quantified germline leakage across 259 sets of whole-genome somatic single nucleotide variant (SNVs) predictions made by 21 teams as part of the ICGC-TCGA DREAM Somatic Mutation Calling Challenge. RESULTS The median somatic SNV prediction set contained 4325 somatic SNVs and leaked one germline polymorphism. The level of germline leakage was inversely correlated with somatic SNV prediction accuracy and positively correlated with the amount of infiltrating normal cells. The specific germline variants leaked differed by tumour and algorithm. To aid in quantitation and correction of leakage, we created a tool, called GermlineFilter, for use in public-facing somatic SNV databases. CONCLUSIONS The potential for patient re-identification from leaked germline variants in somatic SNV predictions has led to divergent open data access policies, based on different assessments of the risks. Indeed, a single, well-publicized re-identification event could reshape public perceptions of the values of genomic data sharing. We find that modern somatic SNV prediction pipelines have low germline-leakage rates, which can be further reduced, especially for cloud-sharing, using pre-filtering software.
Collapse
Affiliation(s)
- Dorota H. Sendorek
- Informatics & Biocomputing Program, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, Ontario M5G 0A3 Canada
| | - Cristian Caloian
- Informatics & Biocomputing Program, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, Ontario M5G 0A3 Canada
| | - Kyle Ellrott
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA USA
- Computational Biology Program, Oregon Health & Science University, Portland, OR USA
| | | | - Takafumi N. Yamaguchi
- Informatics & Biocomputing Program, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, Ontario M5G 0A3 Canada
| | - Adam D. Ewing
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA USA
- Mater Research Institute, University of Queensland, Woolloongabba, Queensland Australia
| | - Kathleen E. Houlahan
- Informatics & Biocomputing Program, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, Ontario M5G 0A3 Canada
| | | | - Adam A. Margolin
- Sage Bionetworks, Seattle, WA USA
- Computational Biology Program, Oregon Health & Science University, Portland, OR USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR USA
| | - Joshua M. Stuart
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA USA
| | - Paul C. Boutros
- Informatics & Biocomputing Program, Ontario Institute for Cancer Research, 661 University Avenue, Suite 510, Toronto, Ontario M5G 0A3 Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario Canada
| |
Collapse
|
31
|
Kytola V, Topaloglu U, Miller LD, Bitting RL, Goodman MM, D`Agostino RB, Desnoyers RJ, Albright C, Yacoub G, Qasem SA, DeYoung B, Thorsson V, Shmulevich I, Yang M, Shcherban A, Pagni M, Liu L, Nykter M, Chen K, Hawkins GA, Grant SC, Petty WJ, Alistar AT, Levine EA, Staren ED, Langefeld CD, Miller V, Singal G, Petro RM, Robinson M, Blackstock W, Powell BL, Wagner LI, Foley KL, Abraham E, Pasche B, Zhang W. Mutational Landscapes of Smoking-Related Cancers in Caucasians and African Americans: Precision Oncology Perspectives at Wake Forest Baptist Comprehensive Cancer Center. Am J Cancer Res 2017; 7:2914-2923. [PMID: 28824725 PMCID: PMC5562225 DOI: 10.7150/thno.20355] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 04/21/2017] [Indexed: 12/17/2022] Open
Abstract
Background: Cancers related to tobacco use and African-American ancestry are under-characterized by genomics. This gap in precision oncology research represents a major challenge in the health disparities in the United States. Methods: The Precision Oncology trial at the Wake Forest Baptist Comprehensive Cancer Center enrolled 431 cancer patients from March 2015 to May 2016. The composition of these patients consists of a high representation of tobacco-related cancers (e.g., lung, colorectal, and bladder) and African-American ancestry (13.5%). Tumors were sequenced to identify mutations to gain insight into genetic alterations associated with smoking and/or African-American ancestry. Results: Tobacco-related cancers exhibit a high mutational load. These tumors are characterized by high-frequency mutations in TP53, DNA damage repair genes (BRCA2 and ATM), and chromatin remodeling genes (the lysine methyltransferases KMT2D or MLL2, and KMT2C or MLL3). These tobacco-related cancers also exhibit augmented tumor heterogeneities. Smoking related genetic mutations were validated by The Cancer Genome Atlas dataset that includes 2,821 cases with known smoking status. The Wake Forest and The Cancer Genome Atlas cohorts (431 and 7,991 cases, respectively) revealed a significantly increased mutation rate in the TP53 gene in the African-American subgroup studied. Both cohorts also revealed 5 genes (e.g. CDK8) significantly amplified in the African-American population. Conclusions: These results provide strong evidence that tobacco is a major cause of genomic instability and heterogeneity in cancer. TP53 mutations and key oncogene amplifications emerge as key factors contributing to cancer outcome disparities among different racial/ethnic groups.
Collapse
|
32
|
Informed consent for next-generation nucleotide sequencing studies: Aiding communication between participants and investigators. J Clin Transl Sci 2017. [PMID: 28649453 PMCID: PMC5471895 DOI: 10.1017/cts.2016.21] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
INTRODUCTION Obtaining informed consent from prospective participants for research studies that include next-generation nucleotide sequencing (NGS) presents significant challenges because of the need to explain all the potential implications of participating, including the possible return of "incidental" findings, in easy-to-understand language. METHODS AND RESULTS After reviewing the consent processes at other institutions, we decided to supplement the protocol-specific informed consent form with the following: (1) a short pamphlet for the prospective participant that includes a series of questions that she or he is encouraged to ask the investigator, and (2) a more detailed companion guide for investigators to help them develop simple-language answers to the questions. Both documents are available to use or modify. CONCLUSIONS We propose an approach to obtaining informed consent for NGS studies that encourages discussion of key issues without creating a complex, comprehensive document for participants; it also maximizes investigator flexibility. We also suggest mechanisms to return restricted information to participants.
Collapse
|
33
|
Goedde LN, Stupiansky NW, Lah M, Quaid KA, Cohen S. Cancer Genetic Counselors’ Current Practices and Attitudes Related to the Use of Tumor Profiling. J Genet Couns 2017; 26:878-886. [DOI: 10.1007/s10897-017-0065-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 01/01/2017] [Indexed: 12/15/2022]
|
34
|
Feliubadaló L, Tonda R, Gausachs M, Trotta JR, Castellanos E, López-Doriga A, Teulé À, Tornero E, Del Valle J, Gel B, Gut M, Pineda M, González S, Menéndez M, Navarro M, Capellá G, Gut I, Serra E, Brunet J, Beltran S, Lázaro C. Benchmarking of Whole Exome Sequencing and Ad Hoc Designed Panels for Genetic Testing of Hereditary Cancer. Sci Rep 2017; 7:37984. [PMID: 28050010 PMCID: PMC5209723 DOI: 10.1038/srep37984] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 11/02/2016] [Indexed: 12/14/2022] Open
Abstract
Next generation sequencing panels have been developed for hereditary cancer, although there is some debate about their cost-effectiveness compared to exome sequencing. The performance of two panels is compared to exome sequencing. Twenty-four patients were selected: ten with identified mutations (control set) and fourteen suspicious of hereditary cancer but with no mutation (discovery set). TruSight Cancer (94 genes) and a custom panel (122 genes) were assessed alongside exome sequencing. Eighty-three genes were targeted by the two panels and exome sequencing. More than 99% of bases had a read depth of over 30x in the panels, whereas exome sequencing covered 94%. Variant calling with standard settings identified the 10 mutations in the control set, with the exception of MSH6 c.255dupC using TruSight Cancer. In the discovery set, 240 unique non-silent coding and canonic splice-site variants were identified in the panel genes, 7 of them putatively pathogenic (in ATM, BARD1, CHEK2, ERCC3, FANCL, FANCM, MSH2). The three approaches identified a similar number of variants in the shared genes. Exomes were more expensive than panels but provided additional data. In terms of cost and depth, panels are a suitable option for genetic diagnostics, although exomes also identify variants in non-targeted genes.
Collapse
Affiliation(s)
- Lídia Feliubadaló
- Hereditary Cancer Program, Joint Program on Hereditary Cancer, Catalan Institute of Oncology, IDIBELL campus in Hospitalet de Llobregat, Catalonia, Spain
| | - Raúl Tonda
- Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain
| | - Mireia Gausachs
- Hereditary Cancer Program, Joint Program on Hereditary Cancer, Catalan Institute of Oncology, IDIBELL campus in Hospitalet de Llobregat, Catalonia, Spain
| | - Jean-Rémi Trotta
- Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain
| | - Elisabeth Castellanos
- Genetic Variation in Cancer Group, Joint Program on Hereditary Cancer, Institut de Medicina Predictiva i Personalitzada del Càncer, Badalona, Catalonia, Spain
| | - Adriana López-Doriga
- Hereditary Cancer Program, Joint Program on Hereditary Cancer, Catalan Institute of Oncology, IDIBELL campus in Hospitalet de Llobregat, Catalonia, Spain
| | - Àlex Teulé
- Hereditary Cancer Program, Joint Program on Hereditary Cancer, Catalan Institute of Oncology, IDIBELL campus in Hospitalet de Llobregat, Catalonia, Spain
| | - Eva Tornero
- Hereditary Cancer Program, Joint Program on Hereditary Cancer, Catalan Institute of Oncology, IDIBELL campus in Hospitalet de Llobregat, Catalonia, Spain
| | - Jesús Del Valle
- Hereditary Cancer Program, Joint Program on Hereditary Cancer, Catalan Institute of Oncology, IDIBELL campus in Hospitalet de Llobregat, Catalonia, Spain
| | - Bernat Gel
- Genetic Variation in Cancer Group, Joint Program on Hereditary Cancer, Institut de Medicina Predictiva i Personalitzada del Càncer, Badalona, Catalonia, Spain
| | - Marta Gut
- Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain
| | - Marta Pineda
- Hereditary Cancer Program, Joint Program on Hereditary Cancer, Catalan Institute of Oncology, IDIBELL campus in Hospitalet de Llobregat, Catalonia, Spain
| | - Sara González
- Hereditary Cancer Program, Joint Program on Hereditary Cancer, Catalan Institute of Oncology, IDIBELL campus in Hospitalet de Llobregat, Catalonia, Spain
| | - Mireia Menéndez
- Hereditary Cancer Program, Joint Program on Hereditary Cancer, Catalan Institute of Oncology, IDIBELL campus in Hospitalet de Llobregat, Catalonia, Spain
| | - Matilde Navarro
- Hereditary Cancer Program, Joint Program on Hereditary Cancer, Catalan Institute of Oncology, IDIBELL campus in Hospitalet de Llobregat, Catalonia, Spain
| | - Gabriel Capellá
- Hereditary Cancer Program, Joint Program on Hereditary Cancer, Catalan Institute of Oncology, IDIBELL campus in Hospitalet de Llobregat, Catalonia, Spain
| | - Ivo Gut
- Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain
| | - Eduard Serra
- Genetic Variation in Cancer Group, Joint Program on Hereditary Cancer, Institut de Medicina Predictiva i Personalitzada del Càncer, Badalona, Catalonia, Spain
| | - Joan Brunet
- Hereditary Cancer Program, Joint Program on Hereditary Cancer, Catalan Institute of Oncology, IdibGi in Girona, Catalonia, Spain
| | - Sergi Beltran
- Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Catalonia, Spain
| | - Conxi Lázaro
- Hereditary Cancer Program, Joint Program on Hereditary Cancer, Catalan Institute of Oncology, IDIBELL campus in Hospitalet de Llobregat, Catalonia, Spain
| |
Collapse
|
35
|
Abstract
Familial pancreatic cancer (FPC) includes those kindreds that contain at least two first-degree relatives with pancreatic ductal adenocarcinoma. At least 12 known hereditary syndromes or genes are associated with increased risk of developing pancreatic cancer, the foremost being BRCA2 and CDKN2A. Research into the identification of mutations in known cancer predisposition genes and through next-generation sequencing has revealed extensive heterogeneity. The development of genetic panel testing has enabled genetic risk assessment and predisposition testing to be routinely offered. Precision oncology has opened the possibility of "incidental" germline mutations that may have implications for family members. However, in both cases, evidence-based recommendations for managing patients and at-risk family members in light of genetic status remain emergent, with current practice based on expert opinion.
Collapse
Affiliation(s)
- Gloria M Petersen
- Department of Health Sciences Research, Mayo Clinic Cancer Center, Rochester, MN.
| |
Collapse
|
36
|
Bourdeaut F. Right to know and right to ignore in paediatric oncogenetics: Identifying biological causes, or seeking for meaning? Eur J Cancer 2016; 63:53-4. [PMID: 27288870 DOI: 10.1016/j.ejca.2016.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 04/24/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Franck Bourdeaut
- Paris Sciences-Lettres Research University, Institut Curie, Departement d'Oncologie Pediatrique-Adolescents et Jeunes adultes, 26 rue d'Ulm, 75005 Paris, France; SiRIC Institut Curie, Laboratoire de Recherche Translationnelle en Oncologie Pediatrique, Institut Curie, 26 rue d'Ulm, 75005 Paris, France; INSERM U830, Genetique et Biologie des Cancers, Institut Curie, 26 rue d'Ulm, 75005 Paris, France.
| |
Collapse
|
37
|
Varga E, Chao EC, Yeager ND. The importance of proper bioinformatics analysis and clinical interpretation of tumor genomic profiling: a case study of undifferentiated sarcoma and a constitutional pathogenic BRCA2 mutation and an MLH1 variant of uncertain significance. Fam Cancer 2016; 14:481-5. [PMID: 25712765 PMCID: PMC4559104 DOI: 10.1007/s10689-015-9790-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Next-generation sequencing (NGS) technology is increasingly utilized to identify therapeutic targets for patients with malignancy. This technology also has the capability to reveal the presence of constitutional genetic alterations, which may have significant implications for patients and their family members. Here we present the case of a 23 year old Caucasian patient with recurrent undifferentiated sarcoma who had NGS-based tumor analysis using an assay which simultaneously analyzed the entire coding sequence of 236 cancer-related genes (3769 exons) plus 47 introns from 19 genes often rearranged or altered in cancer. Pathogenic alterations were reported in tumor as the predicted protein alterations, BRCA2 “R645fs*15″ and MLH1 “E694*”. Because constitutional BRCA2 and MLH1 gene mutations are associated with Hereditary Breast Ovarian Cancer Syndrome (HBOCS) and Lynch syndrome respectively, sequence analysis of DNA isolated from peripheral blood was performed. The presence of the alterations, BRCA2 c.1929delG and MLH1 c.2080G>T, corresponding to the previously reported predicted protein alterations, were confirmed by Sanger sequencing in the constitutional DNA. An additional DNA finding was reported in this analysis, MLH1 c.2081A>C at the neighboring nucleotide. Further evaluation of the family revealed that all alterations were paternally inherited and the two MLH1 substitutions were in cis, more appropriately referred to as MLH1 c.2080_2081delGAinsTC, which is classified as a variant of uncertain significance. This case illustrates important considerations related to appropriate interpretation of NGS tumor results and follow-up of patients with potentially deleterious constitutional alterations.
Collapse
Affiliation(s)
- Elizabeth Varga
- Division of Hematology/Oncology/Bone Marrow Transplant, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH, USA,
| | | | | |
Collapse
|
38
|
Siu LL, Lawler M, Haussler D, Knoppers BM, Lewin J, Vis DJ, Liao RG, Andre F, Banks I, Barrett JC, Caldas C, Camargo AA, Fitzgerald RC, Mao M, Mattison JE, Pao W, Sellers WR, Sullivan P, Teh BT, Ward R, ZenKlusen JC, Sawyers CL, Voest EE. Facilitating a culture of responsible and effective sharing of cancer genome data. Nat Med 2016; 22:464-71. [PMID: 27149219 PMCID: PMC4995884 DOI: 10.1038/nm.4089] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Accepted: 03/21/2016] [Indexed: 12/17/2022]
Abstract
Rapid and affordable tumor molecular profiling has led to an explosion of clinical and genomic data poised to enhance the diagnosis, prognostication and treatment of cancer. A critical point has now been reached at which the analysis and storage of annotated clinical and genomic information in unconnected silos will stall the advancement of precision cancer care. Information systems must be harmonized to overcome the multiple technical and logistical barriers to data sharing. Against this backdrop, the Global Alliance for Genomic Health (GA4GH) was established in 2013 to create a common framework that enables responsible, voluntary and secure sharing of clinical and genomic data. This Perspective from the GA4GH Clinical Working Group Cancer Task Team highlights the data-aggregation challenges faced by the field, suggests potential collaborative solutions and describes how GA4GH can catalyze a harmonized data-sharing culture.
Collapse
Affiliation(s)
- Lillian L. Siu
- Princess Margaret Cancer Centre, University of Toronto, Toronto, Canada
| | - Mark Lawler
- Centre for Cancer Research and Cell Biology, Queen’s University, Belfast, UK
| | - David Haussler
- UC Santa Cruz Genomics Institute, University of California, Santa Cruz, CA, USA
| | | | - Jeremy Lewin
- Princess Margaret Cancer Centre, University of Toronto, Toronto, Canada
| | - Daniel J. Vis
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Rachel G. Liao
- The Global Alliance for Genomics and Health, Toronto, Canada and the Broad Institute, Cambridge, MA, USA
| | - Fabrice Andre
- Gustave Roussy and Université Paris Sud, Villejuif, France
| | - Ian Banks
- Patient’s Advocacy Committee, European Cancer Organization, Brussels, Belgium
| | - J. Carl Barrett
- Translational Sciences, Oncology iMED, AstraZeneca, Waltham, MA, USA
| | | | | | | | - Mao Mao
- Yonsei Cancer Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | | | - William Pao
- Roche Innovation Center Basel, Pharma Research and Early Development, Roche, Basel, Switzerland
| | | | - Patrick Sullivan
- Advocacy for Canadian Children Oncology Network, Vancouver, Canada
| | | | - Robyn Ward
- University of Queensland, St. Lucia, Australia
| | - Jean Claude ZenKlusen
- The Cancer Genome Atlas, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Emile E. Voest
- The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
39
|
Unsolicited findings of next-generation sequencing for tumor analysis within a Dutch consortium: clinical daily practice reconsidered. Eur J Hum Genet 2016; 24:1496-500. [PMID: 27071717 DOI: 10.1038/ejhg.2016.27] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 01/11/2016] [Accepted: 02/10/2016] [Indexed: 12/24/2022] Open
Abstract
Cancer patients participating in studies involving experimental or diagnostic next-generation sequencing (NGS) procedures are confronted with the possibility of unsolicited findings. The Center for Personalized Cancer Treatment (CPCT), a Dutch consortium of cancer centers, is offering centralized large-scale NGS for the discovery of somatic tumor mutations with their germline DNA as reference. The CPCT aims to give all cancer patients with advanced disease stages access to tumor DNA analysis in order to improve selection for experimental therapy. In this article, our experiences at the CPCT will serve as an example to discuss the ethical and practical aspects regarding the management of unsolicited findings in personalized cancer research and treatment. Generic issues, relevant for all researchers in this field are discussed and illustrated by description of three patients faced with an unsolicited DNA finding, while they intended to be candidate for future anticancer treatment by participating in a trial that included NGS of both somatic and germline DNA. As options for DNA analysis expand and costs decrease rapidly, more and more patients are offered large-scale NGS testing. After reviewing current recommendations in literature, we conclude that classical informed consent procedures need to be adapted to become more explicit in asking patients if they want to be informed about unsolicited findings and if so, what level of detail of genetic risk information exactly they want to be returned after the analysis.
Collapse
|
40
|
Kou T, Kanai M, Matsumoto S, Okuno Y, Muto M. The possibility of clinical sequencing in the management of cancer. Jpn J Clin Oncol 2016; 46:399-406. [PMID: 26917600 DOI: 10.1093/jjco/hyw018] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 01/31/2016] [Indexed: 02/07/2023] Open
Abstract
Comprehensive genomic profiling using next-generation sequencing technologies provides insights into understanding the genomic architecture of human cancer. This new understanding of the cancer genome allows us to identify many more genomic alterations occurring within tumors than before, some of which could be potential therapeutic targets through molecular targeted agents. Currently, a large number of molecular targeted agents are being developed, and consequently, cancer treatment is rapidly shifting from empiric therapy employing cytotoxic anticancer drugs to genotype-directed therapy using molecular targeted agents. In current daily clinical practice, hotspot-based single-gene assays that detect RAS mutations in colorectal cancer or EGFR mutations in non-small cell lung cancer are widely used to identify variants. However, it is becoming evident that more comprehensive genomic analysis is crucial in identifying the patient population that may benefit from molecular targeted therapy and the accelerated development of novel drugs for early clinical trials. For these purposes, an increasing number of gene panel-based targeted sequencing is commercially available in clinical practice from sequencing companies. Despite several challenges in implementing this approach, comprehensive genomic profiling and identification of actionable mutations is likely to become one of the standard options in the management of cancer in the near future. The use of clinical sequencing has the potential to usher a new era in precision medicine for cancer diagnosis and treatment. In this review, we discuss the application of comprehensive genomic profiling using next-generation sequencing technologies in clinical oncology and address the current challenges for its implementation.
Collapse
Affiliation(s)
- Tadayuki Kou
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto Department of Clinical Oncology, Kyoto University Hospital Cancer Center, Kyoto
| | - Masashi Kanai
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto Department of Clinical Oncology, Kyoto University Hospital Cancer Center, Kyoto
| | - Shigemi Matsumoto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto Department of Clinical Oncology, Kyoto University Hospital Cancer Center, Kyoto
| | - Yasushi Okuno
- Department of Clinical System Onco-Informatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto Department of Clinical Oncology, Kyoto University Hospital Cancer Center, Kyoto
| |
Collapse
|
41
|
Mak G, Moschetta M, Arkenau HT. Reporting incidental germline variants in the context of day-to-day somatic genomic profiling. Ann Oncol 2016; 27:758-9. [PMID: 26912559 DOI: 10.1093/annonc/mdw078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- G Mak
- Department of Medical Oncology, Sarah Cannon Research Institute UK, London Department of Medical Oncology, University College London Hospitals and Cancer Institute, London, UK
| | - M Moschetta
- Department of Medical Oncology, Sarah Cannon Research Institute UK, London Department of Medical Oncology, University College London Hospitals and Cancer Institute, London, UK
| | - H-T Arkenau
- Department of Medical Oncology, Sarah Cannon Research Institute UK, London Department of Medical Oncology, University College London Hospitals and Cancer Institute, London, UK
| |
Collapse
|
42
|
Boers SN, van Delden JJM, Knoers NV, Bredenoord AL. Postmortem disclosure of genetic information to family members: active or passive? Trends Mol Med 2016; 21:148-53. [PMID: 25743261 DOI: 10.1016/j.molmed.2015.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 12/22/2014] [Accepted: 01/07/2015] [Indexed: 01/19/2023]
Abstract
Advances in next-generation DNA sequencing (NGS) now make it possible, and affordable, to sequence the entire genome of an individual. Routine clinical application is on the horizon. There is a consensus that some subsets of genetic information should be disclosed to patients, but disclosure to their relatives is less consensual. This issue becomes especially salient after a patient's death, when permission can no longer be sought. There has however been little debate on postmortem disclosure. We identify and explain the arguments in favor of and against disclosure of genetic information to the relatives of a deceased patient. We conclude that there are valid reasons to communicate some subsets of genetic information to family members after death, and we propose a passive postmortem disclosure policy.
Collapse
Affiliation(s)
- Sarah N Boers
- Department of Medical Humanities, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Str 6.131, PO Box 85500, 3508 GA Utrecht, The Netherlands.
| | - Johannes J M van Delden
- Department of Medical Humanities, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Str 6.131, PO Box 85500, 3508 GA Utrecht, The Netherlands
| | - Nine V Knoers
- Department of Medical Genetics, University Medical Center Utrecht, KC04.084.2, PO Box 85090, 3508 GA Utrecht, The Netherlands
| | - Annelien L Bredenoord
- Department of Medical Humanities, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Str 6.131, PO Box 85500, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
43
|
Bureau E, Pellegrini I, Noguès C, Lasset C, Julian‐Reynier C. "Maybe they have found something new" participants' views on returning cohort psychosocial survey results. Health Expect 2015; 18:2425-36. [PMID: 24889689 PMCID: PMC5810700 DOI: 10.1111/hex.12211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Although greater attention is currently being paid to participants in research, no studies have dealt so far with the issue of returning aggregate psychosocial results to cohort participants. OBJECTIVE (i) To explore participants' views about disclosure of the aggregate results of a French national psychosocial cohort survey on the epidemiology of preventive behaviour in women from families with a hereditary breast cancer risk. (ii) To assess whether it is worth consulting participants before designing the disclosure process. DESIGN A qualitative study using semi-structured face-to-face interviews and a thematic analysis based on Grounded Theory methods. PARTICIPANTS Nineteen interviews were conducted with cancer-free female BRCA mutation carriers/non-carriers aged 31-79 who had participated in a cohort survey by answering self-administered questionnaires. RESULTS Participants showed considerable interest in the issue of result disclosure. The preferences expressed about disclosure were rarely relevant to the topic investigated, however, as they often focused on medical knowledge about BRCA and not on the psychosocial findings obtained. This confusion may have been due to the participants' experience of the survey procedures, including its longitudinal nature, the occurrence of very few interactions with the investigators and the wide range of topics addressed in the questionnaires. CONCLUSION Investigators should ascertain participants' expectations and preferences by consulting them before disclosing the results obtained. Although the disclosure process may not meet participants' expectations completely, consultation is the key to preventing them from having irrealistic expectations about the information they are going to receive.
Collapse
Affiliation(s)
- Eve Bureau
- INSERMUMR 912MarseilleFrance
- Aix‐Marseille UniversitéUMR 912MarseilleFrance
- IRDUMR 912MarseilleFrance
| | - Isabelle Pellegrini
- INSERMUMR 912MarseilleFrance
- Aix‐Marseille UniversitéUMR 912MarseilleFrance
- IRDUMR 912MarseilleFrance
| | | | | | - Claire Julian‐Reynier
- INSERMUMR 912MarseilleFrance
- Aix‐Marseille UniversitéUMR 912MarseilleFrance
- IRDUMR 912MarseilleFrance
- Institut Paoli‐CalmettesUMR 912MarseilleFrance
| |
Collapse
|
44
|
Hofstatter E, Mehra K, Yushak M, Pusztai L. Tumor profiling and the incidentalome: patient decisions and risks. Future Oncol 2015; 11:3299-305. [DOI: 10.2217/fon.15.260] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
In recent years, the field of oncology has witnessed rapid advancements in genetic sequencing simultaneously with steeply declining costs of sequencing technology. As a result, genomics-driven cancer medicine and the use of tumor profiling are quickly becoming mainstays of cancer therapy. Oncology patients can benefit from tumor profiling by allowing the selection of targeted therapies tailored to their disease. However, it is increasingly recognized that the process of determining a tumor DNA sequence may lead to incidental discovery of underlying germline mutations which can impact other aspects of a patient’s health, and that of their family. How to handle the ‘incidentalome’ has been the subject of recent public debate, yet patient education about the potential risks of tumor profiling remains sparse. Patient perspectives and clinical implications of the tumor incidentalome must be specifically addressed by the oncology community as tumor profiling expands to become a new standard of care.
Collapse
Affiliation(s)
- Erin Hofstatter
- Section of Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Karishma Mehra
- Section of Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Melinda Yushak
- Department of Hematology & Medical Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Lajos Pusztai
- Section of Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
45
|
Fernandez CV, OʼConnell C, Ferguson M, Orr AC, Robitaille JM, Knoppers BM, McMaster CR. Stability of Attitudes to the Ethical Issues Raised by the Return of Incidental Genomic Research Findings in Children: A Follow-Up Study. Public Health Genomics 2015; 18:299-308. [PMID: 26352440 DOI: 10.1159/000439244] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 08/07/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE We explore the stability of parental attitudes to the ethical issues raised by the return of genomic research results. METHODS A 19-item questionnaire was mailed to participants in a large genome research consortium 18 months following a baseline survey. We describe the stability of parental attitudes to (a) sharing of genomic research results, (b) endorsement of children in genomic research, (c) responsibilities of researchers, and (d) responsibilities to extended family. We also explore their experience in receiving results. RESULTS Of 170 original participants, 154 (91%) responded. Most participants expressed positive rights to receive incidental genomic research findings (85%), including when ameliorative therapy was unknown (85%). Only 3% found it acceptable to delegate the decision to return results to an independent committee. Researchers, either with a parent (42%) or physician (17%), were felt to be responsible to convey research results to children when they reach adulthood. Most participants (74%) indicated that results should be shared with potentially affected extended family. These results are very similar to those of the baseline survey. All participants who received genomic results would do so again and reported actions similar to their expressed attitudes. CONCLUSIONS The opinions of parents regarding genomic research remain stable over time. Guidelines on the return of results should incorporate these findings.
Collapse
Affiliation(s)
- Conrad V Fernandez
- Department of Pediatrics, IWK Health Centre and Dalhousie University, Halifax, N.S., Canada
| | | | | | | | | | | | | |
Collapse
|
46
|
Tafe LJ. Targeted Next-Generation Sequencing for Hereditary Cancer Syndromes. J Mol Diagn 2015; 17:472-82. [DOI: 10.1016/j.jmoldx.2015.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 06/09/2015] [Accepted: 06/12/2015] [Indexed: 12/24/2022] Open
|
47
|
Miles G, Rae J, Ramalingam SS, Pfeifer J. Genetic Testing and Tissue Banking for Personalized Oncology: Analytical and Institutional Factors. Semin Oncol 2015; 42:713-23. [PMID: 26433552 DOI: 10.1053/j.seminoncol.2015.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Personalized oncology, or more aptly precision oncogenomics, refers to the identification and implementation of clinically actionable targets tailored to an individual patient's cancer genomic information. Banking of human tissue and other biospecimens establishes a framework to extract and collect the data essential to our understanding of disease pathogenesis and treatment. Cancer cooperative groups in the United States have led the way in establishing robust biospecimen collection mechanisms to facilitate translational research, and combined with technological advances in molecular testing, tissue banking has expanded from its traditional base in academic research and is assuming an increasingly pivotal role in directing the clinical care of cancer patients. Comprehensive screening of tumors by DNA sequencing and the ability to mine and interpret these large data sets from well-organized tissue banks have defined molecular subtypes of cancer. Such stratification by genomic criteria has revolutionized our perspectives on cancer diagnosis and treatment, offering insight into prognosis, progression, and susceptibility or resistance to known therapeutic agents. In turn, this has enabled clinicians to offer treatments tailored to patients that can greatly improve their chances of survival. Unique challenges and opportunities accompany the rapidly evolving interplay between tissue banking and genomic sequencing, and are the driving forces underlying the revolution in precision medicine. Molecular testing and precision medicine clinical trials are now becoming the major thrust behind the cooperative groups' clinical research efforts.
Collapse
Affiliation(s)
- George Miles
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO.
| | - James Rae
- Department of Internal Medicine & Pharmacology, University of Michigan, Ann Arbor, MI
| | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Emory School of Medicine, Winship Cancer Institute, Atlanta, GA
| | - John Pfeifer
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
48
|
Preferences for genetic testing for colorectal cancer within a population-based screening program: a discrete choice experiment. Eur J Hum Genet 2015; 24:361-6. [PMID: 26036860 DOI: 10.1038/ejhg.2015.117] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 04/28/2015] [Accepted: 04/29/2015] [Indexed: 12/17/2022] Open
Abstract
This study explored individuals' preferences for genetic testing for colorectal cancer (CRC) in a screening situation and their willingness to participate in genetic testing for Lynch syndrome, familial adenomatous polyposis (FAP), and familial colorectal cancer (FCC). For that purpose, 532 respondents aged 55-65 years completed a Discrete Choice Experiment. Using panel latent class models, the preferences for two screening situation characteristics (the probability of being genetically predisposed and the probability of developing CRC) and screening test characteristics (the frequency of preventive colonoscopies and CRC survival) were estimated. Based on these preferences, respondents' willingness to participate in the three screening initiatives was estimated. Lower-educated respondents and respondents who express serious anxiety and worries found colonoscopy frequency and the probability of developing CRC relatively more important and survival relatively less important compared with higher-educated respondents and respondents who express no anxiety and worries. These differences in preferences resulted in opposite preferences for participation in FCC and FAP screening. In conclusion, the general population is willing to participate in genetic screening for CRC. If individuals are suspected of genetic or familial CRC, they should at least be informed about their increased risk of being genetically predisposed and about the importance of participating in all preventive follow-up colonoscopies in order to maximize survival.
Collapse
|
49
|
Kuhlen M, Borkhardt A. Cancer susceptibility syndromes in children in the area of broad clinical use of massive parallel sequencing. Eur J Pediatr 2015; 174:987-97. [PMID: 25982339 PMCID: PMC4516864 DOI: 10.1007/s00431-015-2565-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/06/2015] [Accepted: 05/08/2015] [Indexed: 01/23/2023]
Abstract
UNLABELLED Children diagnosed with cancer are considered for inherited cancer susceptibility testing according to well-established clinical criteria. With increasing efforts to personalize cancer medicine, comprehensive genome analyses will find its way into daily clinical routine in pediatric oncology. Whole genome and exome sequencing unavoidably generates incidental findings. The somatic "molecular make-up" of a tumor genome may suggest a germline mutation in a cancer susceptibility syndrome. At least two mechanisms are well-known, (a) chromothripsis (Li-Fraumeni syndrome) and (b) a high total number of mutational events which exceeds that of other samples of the same tumor type (defective DNA mismatch repair). Hence, pediatricians are faced with the fact that genetic events within the tumor genome itself can point toward underlying germline cancer susceptibility. Whenever genetic testing including next-generation sequencing (NGS) is initiated, the pediatrician has to inform about the benefits, risks, and alternatives, discuss the possibility of incidental findings and its disclosure, and to obtain informed consent prior to testing. CONCLUSIONS Genetic testing and translational research in pediatric oncology can incidentally uncover an underlying cancer susceptibility syndrome with implications for the entire family. Pediatricians should therefore increase their awareness of chances and risks that accompany the increasingly wide clinical implementation of NGS platforms.
Collapse
Affiliation(s)
- Michaela Kuhlen
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children's Hospital, Medical Faculty, Heinrich-Heine-University, Moorenstr. 5, 40225, Duesseldorf, Germany,
| | - Arndt Borkhardt
- Department of Pediatric Oncology, Hematology and Clinical Immunology, University Children’s Hospital, Medical Faculty, Heinrich-Heine-University, Moorenstr. 5, 40225 Duesseldorf, Germany
| |
Collapse
|
50
|
Bredenoord AL, Bijlsma RM, van Delden H. Next Generation DNA Sequencing: Always Allow an Opt Out. THE AMERICAN JOURNAL OF BIOETHICS : AJOB 2015; 15:28-30. [PMID: 26147261 DOI: 10.1080/15265161.2015.1039726] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
|