101
|
Ho PJ, Lim EH, Mohamed Ri NKB, Hartman M, Wong FY, Li J. Will Absolute Risk Estimation for Time to Next Screen Work for an Asian Mammography Screening Population? Cancers (Basel) 2023; 15:cancers15092559. [PMID: 37174025 PMCID: PMC10177032 DOI: 10.3390/cancers15092559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Personalized breast cancer risk profiling has the potential to promote shared decision-making and improve compliance with routine screening. We assessed the Gail model's performance in predicting the short-term (2- and 5-year) and the long-term (10- and 15-year) absolute risks in 28,234 asymptomatic Asian women. Absolute risks were calculated using different relative risk estimates and Breast cancer incidence and mortality rates (White, Asian-American, or the Singapore Asian population). Using linear models, we tested the association of absolute risk and age at breast cancer occurrence. Model discrimination was moderate (AUC range: 0.580-0.628). Calibration was better for longer-term prediction horizons (E/Olong-term ranges: 0.86-1.71; E/Oshort-term ranges:1.24-3.36). Subgroup analyses show that the model underestimates risk in women with breast cancer family history, positive recall status, and prior breast biopsy, and overestimates risk in underweight women. The Gail model absolute risk does not predict the age of breast cancer occurrence. Breast cancer risk prediction tools performed better with population-specific parameters. Two-year absolute risk estimation is attractive for breast cancer screening programs, but the models tested are not suitable for identifying Asian women at increased risk within this short interval.
Collapse
Affiliation(s)
- Peh Joo Ho
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore 119228, Singapore
| | - Elaine Hsuen Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 168583, Singapore
| | - Nur Khaliesah Binte Mohamed Ri
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
| | - Mikael Hartman
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore 117549, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore 119228, Singapore
- Department of Surgery, University Surgical Cluster, National University Hospital, Singapore 119228, Singapore
| | - Fuh Yong Wong
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore 168583, Singapore
| | - Jingmei Li
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore 119228, Singapore
| |
Collapse
|
102
|
Lopes Cardozo JM, Andrulis IL, Bojesen SE, Dörk T, Eccles DM, Fasching PA, Hooning MJ, Keeman R, Nevanlinna H, Rutgers EJ, Easton DF, Hall P, Pharoah PD, van 't Veer LJ, Schmidt MK. Associations of a Breast Cancer Polygenic Risk Score With Tumor Characteristics and Survival. J Clin Oncol 2023; 41:1849-1863. [PMID: 36689693 PMCID: PMC10082287 DOI: 10.1200/jco.22.01978] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/25/2022] [Accepted: 12/16/2022] [Indexed: 01/24/2023] Open
Abstract
PURPOSE A polygenic risk score (PRS) consisting of 313 common genetic variants (PRS313) is associated with risk of breast cancer and contralateral breast cancer. This study aimed to evaluate the association of the PRS313 with clinicopathologic characteristics of, and survival following, breast cancer. METHODS Women with invasive breast cancer were included, 98,397 of European ancestry and 12,920 of Asian ancestry, from the Breast Cancer Association Consortium (BCAC), and 683 women from the European MINDACT trial. Associations between PRS313 and clinicopathologic characteristics, including the 70-gene signature for MINDACT, were evaluated using logistic regression analyses. Associations of PRS313 (continuous, per standard deviation) with overall survival (OS) and breast cancer-specific survival (BCSS) were evaluated with Cox regression, adjusted for clinicopathologic characteristics and treatment. RESULTS The PRS313 was associated with more favorable tumor characteristics. In BCAC, increasing PRS313 was associated with lower grade, hormone receptor-positive status, and smaller tumor size. In MINDACT, PRS313 was associated with a low risk 70-gene signature. In European women from BCAC, higher PRS313 was associated with better OS and BCSS: hazard ratio (HR) 0.96 (95% CI, 0.94 to 0.97) and 0.96 (95% CI, 0.94 to 0.98), but the association disappeared after adjustment for clinicopathologic characteristics (and treatment): OS HR, 1.01 (95% CI, 0.98 to 1.05) and BCSS HR, 1.02 (95% CI, 0.98 to 1.07). The results in MINDACT and Asian women from BCAC were consistent. CONCLUSION An increased PRS313 is associated with favorable tumor characteristics, but is not independently associated with prognosis. Thus, PRS313 has no role in the clinical management of primary breast cancer at the time of diagnosis. Nevertheless, breast cancer mortality rates will be higher for women with higher PRS313 as increasing PRS313 is associated with an increased risk of disease. This information is crucial for modeling effective stratified screening programs.
Collapse
Affiliation(s)
- Josephine M.N. Lopes Cardozo
- Department of Surgery, The Netherlands Cancer Institute—Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
- European Organisation for Research and Treatment of Cancer Headquarters, Brussels, Belgium
| | - Irene L. Andrulis
- Fred A. Litwin Center for Cancer Genetics, Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Stig E. Bojesen
- Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thilo Dörk
- Gynaecology Research Unit, Hannover Medical School, Hannover, Germany
| | - Diana M. Eccles
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Peter A. Fasching
- Department of Gynecology and Obstetricss, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| | - Maartje J. Hooning
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Renske Keeman
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Heli Nevanlinna
- Department of Obstetrics and Gynecology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Emiel J.T. Rutgers
- Department of Surgery, The Netherlands Cancer Institute—Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Douglas F. Easton
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Södersjukhuset, Stockholm, Sweden
| | - Paul D.P. Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Laura J. van 't Veer
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA
| | - Marjanka K. Schmidt
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Division of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute—Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
103
|
Thompson HJ, Lutsiv T. Natural Products in Precision Oncology: Plant-Based Small Molecule Inhibitors of Protein Kinases for Cancer Chemoprevention. Nutrients 2023; 15:nu15051192. [PMID: 36904191 PMCID: PMC10005680 DOI: 10.3390/nu15051192] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/17/2023] [Accepted: 02/25/2023] [Indexed: 03/02/2023] Open
Abstract
Striking progress is being made in cancer treatment by using small molecule inhibitors of specific protein kinases that are products of genes recognized as drivers for a specific type of cancer. However, the cost of newly developed drugs is high, and these pharmaceuticals are neither affordable nor accessible in most parts of the world. Accordingly, this narrative review aims to probe how these recent successes in cancer treatment can be reverse-engineered into affordable and accessible approaches for the global community. This challenge is addressed through the lens of cancer chemoprevention, defined as using pharmacological agents of natural or synthetic origin to impede, arrest, or reverse carcinogenesis at any stage in the disease process. In this regard, prevention refers to reducing cancer-related deaths. Recognizing the clinical successes and limitations of protein kinase inhibitor treatment strategies, the disciplines of pharmacognosy and chemotaxonomy are juxtaposed with current efforts to exploit the cancer kinome to describe a conceptual framework for developing a natural product-based approach for precision oncology.
Collapse
Affiliation(s)
- Henry J. Thompson
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO 80523, USA
- Correspondence: ; Tel.: +1-970-491-7748
| | - Tymofiy Lutsiv
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO 80523, USA
- Graduate Program in Cell & Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
104
|
Taylor LC, Law K, Hutchinson A, Dennison RA, Usher-Smith JA. Acceptability of risk stratification within population-based cancer screening from the perspective of healthcare professionals: A mixed methods systematic review and recommendations to support implementation. PLoS One 2023; 18:e0279201. [PMID: 36827432 PMCID: PMC9956883 DOI: 10.1371/journal.pone.0279201] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/01/2022] [Indexed: 02/26/2023] Open
Abstract
BACKGROUND Introduction of risk stratification within population-based cancer screening programmes has the potential to optimise resource allocation by targeting screening towards members of the population who will benefit from it most. Endorsement from healthcare professionals is necessary to facilitate successful development and implementation of risk-stratified interventions. Therefore, this review aims to explore whether using risk stratification within population-based cancer screening programmes is acceptable to healthcare professionals and to identify any requirements for successful implementation. METHODS We searched four electronic databases from January 2010 to October 2021 for quantitative, qualitative, or primary mixed methods studies reporting healthcare professional and/or other stakeholder opinions on acceptability of risk-stratified population-based cancer screening. Quality of the included studies was assessed using the Mixed Methods Appraisal Tool. Data were analysed using the Joanna Briggs Institute convergent integrated approach to mixed methods analysis and mapped onto the Consolidated Framework for Implementation Research using a 'best fit' approach. PROSPERO record CRD42021286667. RESULTS A total of 12,039 papers were identified through the literature search and seven papers were included in the review, six in the context of breast cancer screening and one considering screening for ovarian cancer. Risk stratification was broadly considered acceptable, with the findings covering all five domains of the framework: intervention characteristics, outer setting, inner setting, characteristics of individuals, and process. Across these five domains, key areas that were identified as needing further consideration to support implementation were: a need for greater evidence, particularly for de-intensifying screening; resource limitations; need for staff training and clear communication; and the importance of public involvement. CONCLUSIONS Risk stratification of population-based cancer screening programmes is largely acceptable to healthcare professionals, but support and training will be required to successfully facilitate implementation. Future research should focus on strengthening the evidence base for risk stratification, particularly in relation to reducing screening frequency among low-risk cohorts and the acceptability of this approach across different cancer types.
Collapse
Affiliation(s)
- Lily C. Taylor
- The Primary Care Unit, Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Katie Law
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Alison Hutchinson
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Rebecca A. Dennison
- The Primary Care Unit, Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Juliet A. Usher-Smith
- The Primary Care Unit, Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
105
|
Zhang X, Huang J, Wang J, Li Y, Hu G, Li H. Circ_0001667 accelerates breast cancer proliferation and angiogenesis through regulating CXCL10 expression by sponging miR-6838-5p. Thorac Cancer 2023; 14:881-892. [PMID: 36811283 PMCID: PMC10067355 DOI: 10.1111/1759-7714.14820] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND An increasing number of circular RNAs (circRNAs) have been shown to play an important role in the tumorigenesis of breast cancer. The aim of this study was to investigate the expression and function of circ_0001667 and its potential molecular mechanisms in breast cancer. METHODS The expression levels of circ_0001667, miR-6838-5p and CXC chemokine ligand 10 (CXCL10) in breast cancer tissues and cells were detected by quantitative real-time PCR. Cell counting kit-8 assay, EdU assay, flow cytometry, colony formation and tube formation assays were used to detect cell proliferation and angiogenesis. The binding relationship between miR-6838-5p and circ_0001667 or CXCL10 was predicted using the starBase3.0 database and verified by dual-luciferase reporter gene assay, RIP and RNA pulldown. Animal experiments were performed to assess the function of circ_0001667 knockdown on breast cancer tumor growth. RESULTS Circ_0001667 was highly expressed in breast cancer tissues and cells, and its knockdown inhibited proliferation and angiogenesis of breast cancer cells. Circ_0001667 sponged miR-6838-5p, and inhibition of miR-6838-5p reversed the inhibitory effect of silencing circ_0001667 on proliferation and angiogenesis of breast cancer cells. MiR-6838-5p targeted CXCL10, and overexpression of CXCL10 reverses the effect of miR-6838-5p overexpression on breast cancer cell proliferation and angiogenesis. In addition, circ_0001667 interference also reduced breast cancer tumor growth in vivo. CONCLUSION Circ_0001667 is involved in breast cancer cell proliferation and angiogenesis through regulation of the miR-6838-5p/CXCL10 axis.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Traditional Chinese Medicine, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiami Huang
- Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiayun Wang
- Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yongheng Li
- Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guohua Hu
- Department of Gynecology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - He Li
- Department of Traditional Chinese Medicine, Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
106
|
Heine J, Fowler EE, Weinfurtner RJ, Hume E, Tworoger SS. Breast Density Analysis Using Digital Breast Tomosynthesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.10.527911. [PMID: 36824710 PMCID: PMC9948963 DOI: 10.1101/2023.02.10.527911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
We evaluated an automated percentage of breast density (BD) technique (PDa) with digital breast tomosynthesis (DBT) data. The approach is based on the wavelet expansion followed by analyzing signal dependent noise. Several measures were investigated as risk factors: normalized volumetric; total dense volume; average of the DBT slices (slice-mean); a two-dimensional (2D) metric applied to the synthetic images; and the mean and standard deviations of the pixel values. Volumetric measures were derived theoretically, and PDa was modeled as a function of compressed breast thickness. An alternative method for constructing synthetic 2D mammograms was investigated using the volume results. A matched case-control study (n = 426 pairs) was analyzed. Conditional logistic regression modeling, controlling body mass index and ethnicity, was used to estimate odds ratios (ORs) for each measure with 95% confidence intervals provided parenthetically. There were several significant findings: volumetric measure [OR = 1.43 (1.18, 1.72)], which produced an identical OR as the slice-mean measure as predicted; [OR =1.44 (1.18, 1.75)] when applied to the synthetic images; and mean of the pixel values (volume or 2D synthetic) [ORs ~ 1.31 (1.09, 1.57)]. PDa was modeled as 2nd degree polynomial (concave-down): its maximum value occurred at 0.41×(compressed breast thickness), which was similar across case-control groups, and was significant from this position [OR = 1.47 (1.21, 1.78)]. A standardized 2D synthetic image was produced, where each pixel value represents the percentage of BD above its location. The significant findings indicate the validity of the technique. Derivations supported by empirical analyses produced a new synthetic 2D standardized image technique. Ancillary to the objectives, the results provide evidence for understanding the percentage of BD measure applied to 2D mammograms. Notwithstanding the findings, the study design provides a template for investigating other measures such as texture.
Collapse
|
107
|
French DP, McWilliams L, Bowers S, Woof VG, Harrison F, Ruane H, Hendy A, Evans DG. Psychological impact of risk-stratified screening as part of the NHS Breast Screening Programme: multi-site non-randomised comparison of BC-Predict versus usual screening (NCT04359420). Br J Cancer 2023; 128:1548-1558. [PMID: 36774447 PMCID: PMC9922101 DOI: 10.1038/s41416-023-02156-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 01/08/2023] [Accepted: 01/11/2023] [Indexed: 02/13/2023] Open
Abstract
BACKGROUND Adding risk stratification to standard screening via the NHS Breast Screening Programme (NHSBSP) allows women at higher risk to be offered additional prevention and screening options. It may, however, introduce new harms such as increasing cancer worry. The present study aimed to assess whether there were differences in self-reported harms and benefits between women offered risk stratification (BC-Predict) compared to women offered standard NHSBSP, controlling for baseline values. METHODS As part of the larger PROCAS2 study (NCT04359420), 5901 women were offered standard NHSBSP or BC-Predict at the invitation to NHSBSP. Women who took up BC-Predict received 10-year risk estimates: "high" (≥8%), "above average (moderate)" (5-7.99%), "average" (2-4.99%) or "below average (low)" (<2%) risk. A subset of 662 women completed questionnaires at baseline and at 3 months (n = 511) and 6 months (n = 473). RESULTS State anxiety and cancer worry scores were low with no differences between women offered BC-Predict or NHSBSP. Women offered BC-Predict and informed of being at higher risk reported higher risk perceptions and cancer worry than other women, but without reaching clinical levels. CONCLUSIONS Concerns that risk-stratified screening will produce harm due to increases in general anxiety or cancer worry are unfounded, even for women informed that they are at high risk.
Collapse
Affiliation(s)
- David P. French
- grid.5379.80000000121662407Manchester Centre of Health Psychology, Division of Psychology and Mental Health, School of Health Sciences, University of Manchester, Coupland Street, Manchester, M13 9PL England ,grid.498924.a0000 0004 0430 9101NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, England ,grid.5379.80000000121662407Manchester Breast Centre, Manchester Cancer Research Centre, University of Manchester, 555 Wilmslow Rd, Manchester, M20 4GJ England
| | - Lorna McWilliams
- grid.5379.80000000121662407Manchester Centre of Health Psychology, Division of Psychology and Mental Health, School of Health Sciences, University of Manchester, Coupland Street, Manchester, M13 9PL England ,grid.498924.a0000 0004 0430 9101NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, England
| | - Sarah Bowers
- grid.498924.a0000 0004 0430 9101The Nightingale and Prevent Breast Cancer Centre, Manchester University NHS Foundation Trust, Manchester, M23 9LT England
| | - Victoria G. Woof
- grid.5379.80000000121662407Manchester Centre of Health Psychology, Division of Psychology and Mental Health, School of Health Sciences, University of Manchester, Coupland Street, Manchester, M13 9PL England
| | | | - Helen Ruane
- grid.498924.a0000 0004 0430 9101The Nightingale and Prevent Breast Cancer Centre, Manchester University NHS Foundation Trust, Manchester, M23 9LT England
| | - Alice Hendy
- grid.498924.a0000 0004 0430 9101The Nightingale and Prevent Breast Cancer Centre, Manchester University NHS Foundation Trust, Manchester, M23 9LT England
| | - D. Gareth Evans
- grid.498924.a0000 0004 0430 9101NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, England ,grid.5379.80000000121662407Manchester Breast Centre, Manchester Cancer Research Centre, University of Manchester, 555 Wilmslow Rd, Manchester, M20 4GJ England ,grid.498924.a0000 0004 0430 9101The Nightingale and Prevent Breast Cancer Centre, Manchester University NHS Foundation Trust, Manchester, M23 9LT England ,grid.5379.80000000121662407Genomic Medicine, Division of Evolution and Genomic Sciences, The University of Manchester, St Mary’s Hospital, Manchester University NHS Foundation Trust, Manchester, M13 9WL England
| |
Collapse
|
108
|
Bertozzi S, Londero AP, Xholli A, Azioni G, Di Vora R, Paudice M, Bucimazza I, Cedolini C, Cagnacci A. Risk-Reducing Breast and Gynecological Surgery for BRCA Mutation Carriers: A Narrative Review. J Clin Med 2023; 12:jcm12041422. [PMID: 36835955 PMCID: PMC9967164 DOI: 10.3390/jcm12041422] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023] Open
Abstract
This narrative review aims to clarify the role of breast and gynecological risk-reduction surgery in BRCA mutation carriers. We examine the indications, contraindications, complications, technical aspects, timing, economic impact, ethical issues, and prognostic benefits of the most common prophylactic surgical options from the perspectives of a breast surgeon and a gynecologist. A comprehensive literature review was conducted using the PubMed/Medline, Scopus, and EMBASE databases. The databases were explored from their inceptions to August 2022. Three independent reviewers screened the items and selected those most relevant to this review's scope. BRCA1/2 mutation carriers are significantly more likely to develop breast, ovarian, and serous endometrial cancer. Because of the Angelina effect, there has been a significant increase in bilateral risk-reducing mastectomy (BRRM) since 2013. BRRM and risk-reducing salpingo-oophorectomy (RRSO) significantly reduce the risk of developing breast and ovarian cancer. RRSO has significant side effects, including an impact on fertility and early menopause (i.e., vasomotor symptoms, cardiovascular disease, osteoporosis, cognitive impairment, and sexual dysfunction). Hormonal therapy can help with these symptoms. Because of the lower risk of developing breast cancer in the residual mammary gland tissue after BRRM, estrogen-only treatments have an advantage over an estrogen/progesterone combined treatment. Risk-reducing hysterectomy allows for estrogen-only treatments and lowers the risk of endometrial cancer. Although prophylactic surgery reduces the cancer risk, it has disadvantages associated with early menopause. A multidisciplinary team must carefully inform the woman who chooses this path of the broad spectrum of implications, from cancer risk reduction to hormonal therapies.
Collapse
Affiliation(s)
- Serena Bertozzi
- Breast Unit, University Hospital of Udine, 33100 Udine, UD, Italy
- Ennergi Research (Non-Profit Organisation), 33050 Lestizza, UD, Italy
| | - Ambrogio P. Londero
- Ennergi Research (Non-Profit Organisation), 33050 Lestizza, UD, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Infant Health, University of Genoa, 16132 Genova, GE, Italy
- Correspondence:
| | - Anjeza Xholli
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale San Martino, 16132 Genoa, GE, Italy
| | - Guglielmo Azioni
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale San Martino, 16132 Genoa, GE, Italy
| | - Roberta Di Vora
- Breast Unit, University Hospital of Udine, 33100 Udine, UD, Italy
| | - Michele Paudice
- Anatomic Pathology Unit, Department of Surgical Sciences, and Integrated Diagnostics (DISC), University of Genoa, 16132 Genoa, GE, Italy
- Anatomic Pathology Unit, IRCCS Ospedale San Martino, 16132 Genoa, GE, Italy
| | - Ines Bucimazza
- Department of Surgery, Nelson R. Mandela School of Medicine, University of KwaZulu Natal, Durban 4001, South Africa
| | - Carla Cedolini
- Breast Unit, University Hospital of Udine, 33100 Udine, UD, Italy
- Ennergi Research (Non-Profit Organisation), 33050 Lestizza, UD, Italy
| | - Angelo Cagnacci
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Infant Health, University of Genoa, 16132 Genova, GE, Italy
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale San Martino, 16132 Genoa, GE, Italy
| |
Collapse
|
109
|
Zhao R, Wang W, Pan L, Lv X, He Y, Lian W, Ma Y, Zhang X, Yu R, Zhao S, Guo X, Huang T, Peng M. The prognostic value and response to immunotherapy of immunogenic cell death-associated genes in breast cancer. Front Oncol 2023; 13:1047973. [PMID: 36845750 PMCID: PMC9948621 DOI: 10.3389/fonc.2023.1047973] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/25/2023] [Indexed: 02/11/2023] Open
Abstract
Breast cancer (BRCA) remains the most prevalent cancer worldwide and the tumor microenvironment (TME) has been discovered to exert a wide influence on the overall survival and therapeutic response. Numerous lines of evidence reported that the effects of immunotherapy of BRCA were manipulated by TME. Immunogenic cell death (ICD) is a form of regulated cell death (RCD) that is capable of fueling adaptive immune responses and aberrant expression of ICD-related genes (ICDRGs) can govern the TME system by emitting danger signals or damage-associated molecular patterns (DAMPs). In the current study, we obtained 34 key ICDRGs in BRCA. Subsequently, using the transcriptome data of BRCA from the TCGA database, we constructed a risk signature based on 6 vital ICDRGs, which had a good performance in predicting the overall survival of BRCA patients. We also examined the efficacy of our risk signature in the validation dataset (GSE20711) in the GEO database and it performed excellently. According to the risk model, patients with BRCA were divided into high-risk and low-risk groups. Also, the unique immune characteristics and TME between the two subgroups and 10 promising small molecule drugs targeting BRCA patients with different ICDRGs risk have been investigated. The low-risk group had good immunity indicated by T cell infiltration and high immune checkpoint expression. Moreover, the BRCA samples could be divided into three immune subtypes according to immune response severity (ISA, ISB, and ISC). ISA and ISB predominated in the low-risk group and patients in the low-risk group exhibited a more vigorous immune response. In conclusion, we developed an ICDRGs-based risk signature that can predict the prognosis of BRCA patients and offer a novel therapeutic strategy for immunotherapy, which would be of great significance in the BRCA clinical setting.
Collapse
Affiliation(s)
- Rongling Zhao
- Department of Clinical Laboratory, Henan No.3 Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Wenkang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Limin Pan
- Department of Breast Surgery, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Xuefeng Lv
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi He
- Department of Mini-Invasive Spinal Surgery, Henan No.3 Provincial People’s Hospital, Zhengzhou, China
| | - Wenping Lian
- Department of Clinical Laboratory, Henan No.3 Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Yajie Ma
- Department of Medical Affair, Henan No.3 Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Xinyu Zhang
- Department of Medical Affair, Henan No.3 Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Ruijing Yu
- Department of Clinical Laboratory, Henan No.3 Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Shuai Zhao
- Department of Clinical Laboratory, Henan No.3 Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Xiaona Guo
- Medical School, Huanghe Science and Technology University, Zhengzhou, Henan, China
| | - Tao Huang
- Medical School, Huanghe Science and Technology University, Zhengzhou, Henan, China,*Correspondence: Mengle Peng, ; Tao Huang,
| | - Mengle Peng
- Department of Clinical Laboratory, Henan No.3 Provincial People’s Hospital, Zhengzhou, Henan, China,*Correspondence: Mengle Peng, ; Tao Huang,
| |
Collapse
|
110
|
Ha M, Ngaage LM, Finkelstein ER, Klein M, Yanga A, Colohan SM, Nurudeen SM, Terhune JH, Slezak S, Rasko YM. Insurance Coverage of Prophylactic Mastectomies: A National Review of the United States. Clin Breast Cancer 2023; 23:211-218. [PMID: 36588087 DOI: 10.1016/j.clbc.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Breast cancer is associated with a multitude of risk factors, such as genetic predisposition and mutations, family history, personal medical history, or previous radiotherapy. A prophylactic mastectomy (PM) may be considered a suitable risk-reducing procedure in some cases. However, there are significant discrepancies between national society recommendations and insurance company requirements for PM. MATERIALS AND METHODS The authors conducted a cross-sectional analysis of insurance policies for a PM. One-hundred companies were selected based on the greatest state enrolment and market share. Their policies were identified through a Web-based search and telephone interviews, and their medical necessity criteria were extracted. RESULTS Preauthorized coverage of PMs was provided by 39% of insurance policies (n = 39) and 5 indications were identified. There was consensus amongst these policies to cover a PM for BRCA1/2 mutations (n = 39, 100%), but was more variable for other genetic mutations (15%-90%). Coverage of PM for the remaining indications varied among insurers: previous radiotherapy (92%), pathological changes in the breast (3%-92%), personal history of cancer (64%) and family history risk factors (39%-51%). CONCLUSION There is a marked level of variability in both the indications and medical necessity criteria for PM insurance policies. The decision to undergo a PM must be carefully considered with a patient's care team and should not be affected by insurance coverage status.
Collapse
Affiliation(s)
- Michael Ha
- Division of Plastic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Ledibabari M Ngaage
- Division of Plastic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD; Department of Plastic and Reconstructive Surgery, Johns Hopkins University, Baltimore, MD
| | - Emily R Finkelstein
- Division of Plastic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD; Division of Plastic and Reconstructive Surgery, University of Miami Miller School of Medicine, Miami, FL.
| | - Marissa Klein
- Division of Plastic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Annie Yanga
- Division of Plastic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Shannon M Colohan
- Division of Plastic Surgery, Department of Surgery, University of Washington, Seattle, WA
| | - Suliat M Nurudeen
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Julia H Terhune
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Sheri Slezak
- Division of Plastic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Yvonne M Rasko
- Division of Plastic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
111
|
Chang Y, Huang Z, Quan H, Li H, Yang S, Song Y, Wang J, Yuan J, Wu C. Construction of a DNA damage repair gene signature for predicting prognosis and immune response in breast cancer. Front Oncol 2023; 12:1085632. [PMID: 36713553 PMCID: PMC9875088 DOI: 10.3389/fonc.2022.1085632] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/13/2022] [Indexed: 01/13/2023] Open
Abstract
DNA damage repair (DDR) genes are involved in developing breast cancer. Recently, a targeted therapeutic strategy through DNA repair machinery, including PARPi, has initially shown broad development and application prospects in breast cancer therapy. However, few studies that focused on the correlation between the expression level of DNA repair genes, prognosis, and immune response in breast cancer patients have been recently conducted. Herein, we focused on identifying differentially expressed DNA repair genes (DEGs) in breast cancer specimens and normal samples using the Wilcoxon rank-sum test. Biofunction enrichment analysis was performed with DEGs using the R software "cluster Profiler" package. DNA repair genes were involved in multivariate and univariate Cox regression analyses. After the optimization by AIC value, 11 DNA repair genes were sorted as prognostic DNA repair genes for breast cancer patients to calculate risk scores. Simultaneously, a nomogram was used to represent the prognostic model, which was validated using a calibration curve and C-index. Single-sample gene set enrichment analysis (ssGSEA), CIBERSORT algorithms, and ESTIMATE scores were applied to evaluate the immune filtration of tumor samples. Subsequently, anticarcinogen sensitivity analysis was performed using the R software "pRRophetic" package. Unsupervised clustering was used to excavate the correlation between the expression level of prognostic-significant DNA repair genes and clinical features. In summary, 56 DEGs were sorted, and their potential enriched biofunction pathways were revealed. In total, 11 DNA repair genes (UBE2A, RBBP8, RAD50 , FAAP20, RPA3, ENDOV, DDB2, UBE2V2, MRE11 , RRM2B, and PARP3 ) were preserved as prognostic genes to estimate risk score, which was applied to establish the prognostic model and stratified breast cancer patients into two groups with high or low risk. The calibration curve and C-index indicated that they reliably predicted the survival of breast cancer patients. Immune filtration analysis, anticarcinogen sensitivity analysis, and unsupervised clustering were applied to reveal the character of DNA repair genes between low- and high-risk groups. We identified 11 prognosis-significant DNA repair genes to establish prediction models and immune responses in breast cancer patients.
Collapse
Affiliation(s)
- Yiming Chang
- Jinzhou Medical University, Shanghai East Hospital, Shanghai, China
| | - Zhiyuan Huang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hong Quan
- Department of Breast Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hui Li
- Department of Gynaecology and Obstetrics, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shuo Yang
- Department of Medical Imaging, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yifei Song
- Department of Biochemistry and Molecular Biology, Tongji University School of Medicine, Shanghai, China
| | - Jian Wang
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, China,*Correspondence: Jian Yuan, ; Chenming Wu, ; Jian Wang,
| | - Jian Yuan
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China,Department of Biochemistry and Molecular Biology, Tongji University School of Medicine, Shanghai, China,Ji’an Hospital, Shanghai East Hospital, Ji’an, China,*Correspondence: Jian Yuan, ; Chenming Wu, ; Jian Wang,
| | - Chenming Wu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China,*Correspondence: Jian Yuan, ; Chenming Wu, ; Jian Wang,
| |
Collapse
|
112
|
Ayana G, Dese K, Dereje Y, Kebede Y, Barki H, Amdissa D, Husen N, Mulugeta F, Habtamu B, Choe SW. Vision-Transformer-Based Transfer Learning for Mammogram Classification. Diagnostics (Basel) 2023; 13:diagnostics13020178. [PMID: 36672988 PMCID: PMC9857963 DOI: 10.3390/diagnostics13020178] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/27/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
Breast mass identification is a crucial procedure during mammogram-based early breast cancer diagnosis. However, it is difficult to determine whether a breast lump is benign or cancerous at early stages. Convolutional neural networks (CNNs) have been used to solve this problem and have provided useful advancements. However, CNNs focus only on a certain portion of the mammogram while ignoring the remaining and present computational complexity because of multiple convolutions. Recently, vision transformers have been developed as a technique to overcome such limitations of CNNs, ensuring better or comparable performance in natural image classification. However, the utility of this technique has not been thoroughly investigated in the medical image domain. In this study, we developed a transfer learning technique based on vision transformers to classify breast mass mammograms. The area under the receiver operating curve of the new model was estimated as 1 ± 0, thus outperforming the CNN-based transfer-learning models and vision transformer models trained from scratch. The technique can, hence, be applied in a clinical setting, to improve the early diagnosis of breast cancer.
Collapse
Affiliation(s)
- Gelan Ayana
- Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39253, Republic of Korea
- School of Biomedical Engineering, Jimma University, Jimma 378, Ethiopia
| | - Kokeb Dese
- School of Biomedical Engineering, Jimma University, Jimma 378, Ethiopia
| | - Yisak Dereje
- Department of Information Engineering, Marche Polytechnic University, 60121 Ancona, Italy
| | - Yonas Kebede
- Biomedical Engineering Unit, Black Lion Hospital, Addis Ababa University, Addis Ababa 1000, Ethiopia
| | - Hika Barki
- Department of Artificial Intelligence Convergence, Pukyong National University, Busan 48513, Republic of Korea
| | - Dechassa Amdissa
- Department of Basic and Applied Science for Engineering, Sapienza University of Rome, 00161 Roma, Italy
| | - Nahimiya Husen
- Department of Bioengineering and Robotics, Campus Bio-Medico University of Rome, 00128 Roma, Italy
| | - Fikadu Mulugeta
- Center of Biomedical Engineering, Addis Ababa Institute of Technology, Addis Ababa University, Addis Ababa 1000, Ethiopia
| | - Bontu Habtamu
- School of Biomedical Engineering, Jimma University, Jimma 378, Ethiopia
| | - Se-Woon Choe
- Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39253, Republic of Korea
- Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39253, Republic of Korea
- Correspondence: ; Tel.: +82-54-478-7781; Fax: +82-54-462-1049
| |
Collapse
|
113
|
Trapani D, Sandoval J, Aliaga PT, Ascione L, Maria Berton Giachetti PP, Curigliano G, Ginsburg O. Screening Programs for Breast Cancer: Toward Individualized, Risk-Adapted Strategies of Early Detection. Cancer Treat Res 2023; 188:63-88. [PMID: 38175342 DOI: 10.1007/978-3-031-33602-7_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Early detection of breast cancer (BC) comprises two approaches: screening of asymptomatic women in a specified target population at risk (usually a target age range for women at average risk), and early diagnosis for women with BC signs and symptoms. Screening for BC is a key health intervention for early detection. While population-based screening programs have been implemented for age-selected women, the pivotal clinical trials have not addressed the global utility nor the improvement of screening performance by utilizing more refined parameters for patient eligibility, such as individualized risk stratification. In addition, with the exception of the subset of women known to carry germline pathogenetic mutations in (high- or moderately-penetrant) cancer predisposition genes, such as BRCA1 and BRCA2, there has been less success in outreach and service provision for the unaffected relatives of women found to carry a high-risk mutation (i.e., "cascade testing") as it is in these individuals for whom such actionable information can result in cancers (and/or cancer deaths) being averted. Moreover, even in the absence of clinical cancer genetics services, as is the case for the immediate and at least near-term in most countries globally, the capacity to stratify the risk of an individual to develop BC has existed for many years, is available for free online at various sites/platforms, and is increasingly being validated for non-Caucasian populations. Ultimately, a precision approach to BC screening is largely missing. In the present chapter, we aim to address the concept of risk-adapted screening of BC, in multiple facets, and understand if there is a value in the implementation of adapted screening strategies in selected women, outside the established screening prescriptions, in the terms of age-range, screening modality and schedules of imaging.
Collapse
Affiliation(s)
- Dario Trapani
- Division of New Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy.
| | - Josè Sandoval
- Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Unit of Population Epidemiology, Division and Department of Primary Care Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Pamela Trillo Aliaga
- Division of New Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Liliana Ascione
- Division of New Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Pier Paolo Maria Berton Giachetti
- Division of New Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | | |
Collapse
|
114
|
Magni V, Cozzi A, Schiaffino S, Colarieti A, Sardanelli F. Artificial intelligence for digital breast tomosynthesis: Impact on diagnostic performance, reading times, and workload in the era of personalized screening. Eur J Radiol 2023; 158:110631. [PMID: 36481480 DOI: 10.1016/j.ejrad.2022.110631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022]
Abstract
The ultimate goals of the application of artificial intelligence (AI) to digital breast tomosynthesis (DBT) are the reduction of reading times, the increase of diagnostic performance, and the reduction of interval cancer rates. In this review, after outlining the journey from computer-aided detection/diagnosis systems to AI applied to digital mammography (DM), we summarize the results of studies where AI was applied to DBT, noting that long-term advantages of DBT screening and its crucial ability to decrease the interval cancer rate are still under scrutiny. AI has shown the capability to overcome some shortcomings of DBT in the screening setting by improving diagnostic performance and by reducing recall rates (from -2 % to -27 %) and reading times (up to -53 %, with an average 20 % reduction), but the ability of AI to reduce interval cancer rates has not yet been clearly investigated. Prospective validation is needed to assess the cost-effectiveness and real-world impact of AI models assisting DBT interpretation, especially in large-scale studies with low breast cancer prevalence. Finally, we focus on the incoming era of personalized and risk-stratified screening that will first see the application of contrast-enhanced breast imaging to screen women with extremely dense breasts. As the diagnostic advantage of DBT over DM was concentrated in this category, we try to understand if the application of AI to DM in the remaining cohorts of women with heterogeneously dense or non-dense breast could close the gap in diagnostic performance between DM and DBT, thus neutralizing the usefulness of AI application to DBT.
Collapse
Affiliation(s)
- Veronica Magni
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Via Luigi Mangiagalli 31, 20133 Milano, Italy.
| | - Andrea Cozzi
- Unit of Radiology, IRCCS Policlinico San Donato, Via Rodolfo Morandi 30, 20097 San Donato Milanese, Italy
| | - Simone Schiaffino
- Unit of Radiology, IRCCS Policlinico San Donato, Via Rodolfo Morandi 30, 20097 San Donato Milanese, Italy
| | - Anna Colarieti
- Unit of Radiology, IRCCS Policlinico San Donato, Via Rodolfo Morandi 30, 20097 San Donato Milanese, Italy
| | - Francesco Sardanelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Via Luigi Mangiagalli 31, 20133 Milano, Italy; Unit of Radiology, IRCCS Policlinico San Donato, Via Rodolfo Morandi 30, 20097 San Donato Milanese, Italy.
| |
Collapse
|
115
|
Anandarajah A, Chen Y, Colditz GA, Hardi A, Stoll C, Jiang S. Studies of parenchymal texture added to mammographic breast density and risk of breast cancer: a systematic review of the methods used in the literature. Breast Cancer Res 2022; 24:101. [PMID: 36585732 PMCID: PMC9805242 DOI: 10.1186/s13058-022-01600-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
This systematic review aimed to assess the methods used to classify mammographic breast parenchymal features in relation to the prediction of future breast cancer. The databases including Medline (Ovid) 1946-, Embase.com 1947-, CINAHL Plus 1937-, Scopus 1823-, Cochrane Library (including CENTRAL), and Clinicaltrials.gov were searched through October 2021 to extract published articles in English describing the relationship of parenchymal texture features with the risk of breast cancer. Twenty-eight articles published since 2016 were included in the final review. The identification of parenchymal texture features varied from using a predefined list to machine-driven identification. A reduction in the number of features chosen for subsequent analysis in relation to cancer incidence then varied across statistical approaches and machine learning methods. The variation in approach and number of features identified for inclusion in analysis precluded generating a quantitative summary or meta-analysis of the value of these features to improve predicting risk of future breast cancers. This updated overview of the state of the art revealed research gaps; based on these, we provide recommendations for future studies using parenchymal features for mammogram images to make use of accumulating image data, and external validation of prediction models that extend to 5 and 10 years to guide clinical risk management. Following these recommendations could enhance the applicability of models, helping improve risk classification and risk prediction for women to tailor screening and prevention strategies to the level of risk.
Collapse
Affiliation(s)
- Akila Anandarajah
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, 660 S Euclid Ave MSC 8100-0094-2200, Saint Louis, MO, 63110, USA
| | - Yongzhen Chen
- Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Graham A Colditz
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, 660 S Euclid Ave MSC 8100-0094-2200, Saint Louis, MO, 63110, USA
| | - Angela Hardi
- Bernard Becker Medical Library, Washington University School of Medicine, MSC 8132-12-01, 660 S Euclid Ave, Saint Louis, MO, 63110, USA
| | - Carolyn Stoll
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, 660 S Euclid Ave MSC 8100-0094-2200, Saint Louis, MO, 63110, USA
| | - Shu Jiang
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, 660 S Euclid Ave MSC 8100-0094-2200, Saint Louis, MO, 63110, USA.
| |
Collapse
|
116
|
Alasmari MM. A Review of Margetuximab-Based Therapies in Patients with HER2-Positive Metastatic Breast Cancer. Cancers (Basel) 2022; 15:cancers15010038. [PMID: 36612034 PMCID: PMC9817862 DOI: 10.3390/cancers15010038] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) is the most commonly diagnosed cancer globally, with high mortality rates. Targeted drug therapies have revolutionized cancer treatment. For example, treatment with human epidermal receptor 2 (HER2) antagonists has markedly improved the prognosis of patients with HER2-positive BC (HER2 + BC). However, HER2+ metastatic BC (MBC) remains prevalent owing to its resistance to conventional anti-HER2 drugs. Therefore, novel agents are needed to overcome the limitations of existing cancer treatments and to enhance the progression-free and overall survival rates. Progress has been made by optimizing the fragment crystallizable (Fc) domain of trastuzumab, an IgG1 monoclonal, chimeric anti-HER2 antibody, to develop margetuximab. The modified Fc domain of margetuximab enhances its binding affinity to CD16A and decreases its binding affinity to CD32B, thereby promoting its antitumor activity. This review summarizes studies on the efficacy of margetuximab, discusses its utility as an anti-HER2 monoclonal antibody drug for the treatment of HER2 + BC, and presents the latest advances in the treatment of BC. This review provides insights into the clinical implication of margetuximab in HER2 + MBC treatment.
Collapse
Affiliation(s)
- Moudi M. Alasmari
- College of Medicine, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), Jeddah 21461, Saudi Arabia;
- King Abdullah International Medical Research Centre (KAIMRC), Jeddah 21423, Saudi Arabia
| |
Collapse
|
117
|
Durham DD, Abraham LA, Roberts MC, Khan CP, Smith RA, Kerlikowske K, Miglioretti DL. Breast cancer incidence among women with a family history of breast cancer by relative's age at diagnosis. Cancer 2022; 128:4232-4240. [PMID: 36262035 PMCID: PMC9712500 DOI: 10.1002/cncr.34365] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/03/2021] [Accepted: 01/07/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Women with a first-degree family history of breast cancer are often advised to begin screening when they are 10 years younger than the age at which their relative was diagnosed. Evidence is lacking to determine how much earlier they should begin. METHODS Using Breast Cancer Surveillance Consortium data on screening mammograms from 1996 to 2016, the authors constructed a cohort of 306,147 women 30-59 years of age with information on first-degree family history of breast cancer and relative's age at diagnosis. The authors compared cumulative 5-year breast cancer incidence among women with and without a first-degree family history of breast by relative's age at diagnosis and by screening age. RESULTS Among 306,147 women included in the study, approximately 11% reported a first-degree family history of breast cancer with 3885 breast cancer cases identified. Women reporting a relative diagnosed between 40 and 49 years and undergoing screening between ages 30 and 39 or 40 and 49 had similar 5-year cumulative incidences of breast cancer (respectively, 18.6/1000; 95% confidence interval [CI], 12.1, 25.7; 18.4/1000; 95% CI, 13.7, 23.5) as women without a family history undergoing screening between 50-59 years of age (18.0/1000; 95% CI, 17.0, 19.1). For relative's diagnosis age from 35 to 45 years of age, initiating screening 5-8 years before diagnosis age resulted in a 5-year cumulative incidence of breast cancer of 15.2/1000, that of an average 50-year-old woman. CONCLUSION Women with a relative diagnosed at or before age 45 may wish to consider, in consultation with their provider, initiating screening 5-8 years earlier than their relative's diagnosis age.
Collapse
Affiliation(s)
- Danielle D. Durham
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Cancer Prevention Fellowship Program, Division of Cancer Prevention, Healthcare Delivery Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland, USA
| | - Linn A. Abraham
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, Washington, USA
| | - Megan C. Roberts
- Division of Pharmaceutical Outcomes and Policy, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, North Carolina, USA
- Cancer Prevention Fellowship Program, Division of Cancer Prevention, Behavioral Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland, USA
| | - Carly P. Khan
- Patient-Centered Outcomes Research Institute, Washington, District of Columbia, USA
- Cancer Prevention Fellowship Program, Division of Cancer Prevention, Community Oncology and Prevention Trials Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland, USA
| | - Robert A. Smith
- Cancer Control Department, American Cancer Society, Atlanta, Georgia, USA
| | - Karla Kerlikowske
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
| | - Diana L. Miglioretti
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, Washington, USA
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, Davis, California, USA
| |
Collapse
|
118
|
Li Y, Feng J, Wang T, Li M, Zhang H, Rong Z, Cheng W, Duan Y, Chen Z, Hu A, Yu T, Zhang J, Shang Y, Zou Y, Ma F, Guo B. Construction of an immunogenic cell death-based risk score prognosis model in breast cancer. Front Genet 2022; 13:1069921. [PMID: 36583019 PMCID: PMC9792780 DOI: 10.3389/fgene.2022.1069921] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Immunogenic cell death (ICD) is a form of regulated cell death that elicits immune response. Common inducers of ICD include cancer chemotherapy and radiation therapy. A better understanding of ICD might contribute to modify the current regimens of anti-cancer therapy, especially immunotherapy. This study aimed to identify ICD-related prognostic gene signatures in breast cancer (BC). An ICD-based gene prognostic signature was developed using Lasso-cox regression and Kaplan-Meier survival analysis based on datasets acquired from the Cancer Genome Atlas and Gene Expression Omnibus. A nomogram model was developed to predict the prognosis of BC patients. Gene Set Enrichment Analysis (GESA) and Gene Set Variation Analysis (GSVA) were used to explore the differentially expressed signaling pathways in high and low-risk groups. CIBERSORT and ESTIMATE algorithms were performed to investigate the difference of immune status in tumor microenvironment of different risk groups. Six genes (CALR, CLEC9A, BAX, TLR4, CXCR3, and PIK3CA) were selected for construction and validation of the prognosis model of BC based on public data. GSEA and GSVA analysis found that immune-related gene sets were enriched in low-risk group. Moreover, immune cell infiltration analysis showed that the immune features of the high-risk group were characterized by higher infiltration of tumor-associated macrophages and a lower proportion of CD8+ T cells, suggesting an immune evasive tumor microenvironment. We constructed and validated an ICD-based gene signature for predicting prognosis of breast cancer patients. Our model provides a tool with good discrimination and calibration abilities to predict the prognosis of BC, especially triple-negative breast cancer (TNBC).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Fei Ma
- *Correspondence: Fei Ma, ; Baoliang Guo,
| | | |
Collapse
|
119
|
Gorman LS, Ruane H, Woof VG, Southworth J, Ulph F, Evans DG, French DP. The co-development of personalised 10-year breast cancer risk communications: a 'think-aloud' study. BMC Cancer 2022; 22:1264. [PMID: 36471302 PMCID: PMC9721070 DOI: 10.1186/s12885-022-10347-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/21/2022] [Indexed: 12/09/2022] Open
Abstract
BACKGROUND Risk stratified breast cancer screening is being considered as a means of improving the balance of benefits and harms of mammography. Stratified screening requires the communication of risk estimates. We aimed to co-develop personalised 10-year breast cancer risk communications for women attending routine mammography. METHODS We conducted think-aloud interviews on prototype breast cancer risk letters and accompanying information leaflets with women receiving breast screening through the UK National Breast Screening Programme. Risk information was redesigned following feedback from 55 women in three iterations. A deductive thematic analysis of participants' speech is presented. RESULTS Overall, participants appreciated receiving their breast cancer risk. Their comments focused on positive framing and presentation of the risk estimate, a desire for detail on the contribution of individual risk factors to overall risk and effective risk management strategies, and clearly signposted support pathways. CONCLUSION Provision of breast cancer risk information should strive to be personal, understandable and meaningful. Risk information should be continually refined to reflect developments in risk management. Receipt of risk via letter is welcomed but concerns remain around the acceptability of informing women at higher risk in this way, highlighting a need for co-development of risk dissemination and support pathways.
Collapse
Affiliation(s)
- Louise S. Gorman
- grid.498924.a0000 0004 0430 9101The Nightingale Centre and Prevent Breast Cancer Centre Research Unit, Manchester University NHS Foundation Trust, Southmoor Road, Manchester, M23 9LT UK
| | - Helen Ruane
- grid.498924.a0000 0004 0430 9101The Nightingale Centre and Prevent Breast Cancer Centre Research Unit, Manchester University NHS Foundation Trust, Southmoor Road, Manchester, M23 9LT UK
| | - Victoria G. Woof
- grid.5379.80000000121662407Manchester Centre for Health Psychology, Division of Psychology & Mental Health, School of Health Sciences, The University of Manchester, MAHSC, Oxford Road, Manchester, M13 9PL UK
| | - Jake Southworth
- grid.498924.a0000 0004 0430 9101The Nightingale Centre and Prevent Breast Cancer Centre Research Unit, Manchester University NHS Foundation Trust, Southmoor Road, Manchester, M23 9LT UK
| | - Fiona Ulph
- grid.5379.80000000121662407Manchester Centre for Health Psychology, Division of Psychology & Mental Health, School of Health Sciences, The University of Manchester, MAHSC, Oxford Road, Manchester, M13 9PL UK
| | - D. Gareth Evans
- grid.498924.a0000 0004 0430 9101The Nightingale Centre and Prevent Breast Cancer Centre Research Unit, Manchester University NHS Foundation Trust, Southmoor Road, Manchester, M23 9LT UK ,grid.498924.a0000 0004 0430 9101Department of Genomic Medicine, Division of Evolution and Genomic Science, MAHSC, University of Manchester, Manchester University NHS Foundation Trust, Oxford Road, M13 9WL, Manchester, UK ,grid.498924.a0000 0004 0430 9101NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, England UK
| | - David P. French
- grid.5379.80000000121662407Manchester Centre for Health Psychology, Division of Psychology & Mental Health, School of Health Sciences, The University of Manchester, MAHSC, Oxford Road, Manchester, M13 9PL UK ,grid.498924.a0000 0004 0430 9101NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester, England UK
| |
Collapse
|
120
|
Lee A, Mavaddat N, Cunningham A, Carver T, Ficorella L, Archer S, Walter FM, Tischkowitz M, Roberts J, Usher-Smith J, Simard J, Schmidt MK, Devilee P, Zadnik V, Jürgens H, Mouret-Fourme E, De Pauw A, Rookus M, Mooij TM, Pharoah PP, Easton DF, Antoniou AC. Enhancing the BOADICEA cancer risk prediction model to incorporate new data on RAD51C, RAD51D, BARD1 updates to tumour pathology and cancer incidence. J Med Genet 2022; 59:1206-1218. [PMID: 36162851 PMCID: PMC9691826 DOI: 10.1136/jmedgenet-2022-108471] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/23/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND BOADICEA (Breast and Ovarian Analysis of Disease Incidence and Carrier Estimation Algorithm) for breast cancer and the epithelial tubo-ovarian cancer (EOC) models included in the CanRisk tool (www.canrisk.org) provide future cancer risks based on pathogenic variants in cancer-susceptibility genes, polygenic risk scores, breast density, questionnaire-based risk factors and family history. Here, we extend the models to include the effects of pathogenic variants in recently established breast cancer and EOC susceptibility genes, up-to-date age-specific pathology distributions and continuous risk factors. METHODS BOADICEA was extended to further incorporate the associations of pathogenic variants in BARD1, RAD51C and RAD51D with breast cancer risk. The EOC model was extended to include the association of PALB2 pathogenic variants with EOC risk. Age-specific distributions of oestrogen-receptor-negative and triple-negative breast cancer status for pathogenic variant carriers in these genes and CHEK2 and ATM were also incorporated. A novel method to include continuous risk factors was developed, exemplified by including adult height as continuous. RESULTS BARD1, RAD51C and RAD51D explain 0.31% of the breast cancer polygenic variance. When incorporated into the multifactorial model, 34%-44% of these carriers would be reclassified to the near-population and 15%-22% to the high-risk categories based on the UK National Institute for Health and Care Excellence guidelines. Under the EOC multifactorial model, 62%, 35% and 3% of PALB2 carriers have lifetime EOC risks of <5%, 5%-10% and >10%, respectively. Including height as continuous, increased the breast cancer relative risk variance from 0.002 to 0.010. CONCLUSIONS These extensions will allow for better personalised risks for BARD1, RAD51C, RAD51D and PALB2 pathogenic variant carriers and more informed choices on screening, prevention, risk factor modification or other risk-reducing options.
Collapse
Affiliation(s)
- Andrew Lee
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Nasim Mavaddat
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Alex Cunningham
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Tim Carver
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Lorenzo Ficorella
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Stephanie Archer
- Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Fiona M Walter
- Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Marc Tischkowitz
- Department of Medical Genetics and National Institute for Health Research, Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Jonathan Roberts
- Department of Medical Genetics and National Institute for Health Research, Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Juliet Usher-Smith
- Primary Care Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Jacques Simard
- Centre Hospitalier Universitaire de Québec-Université Laval Research Center, Université Laval, Quebec, Quebec, Canada
| | - Marjanka K Schmidt
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Peter Devilee
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Vesna Zadnik
- Epidemiology and Cancer Registry, Institute of Oncology, Ljubljana, Slovenia
| | - Hannes Jürgens
- Clinic of Hematology and Oncology, Tartu University Hospital, Tartu, Estonia
| | | | | | - Matti Rookus
- Department of Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Thea M Mooij
- Department of Epidemiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Paul Pd Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Antonis C Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| |
Collapse
|
121
|
Yang X, Eriksson M, Czene K, Lee A, Leslie G, Lush M, Wang J, Dennis J, Dorling L, Carvalho S, Mavaddat N, Simard J, Schmidt MK, Easton DF, Hall P, Antoniou AC. Prospective validation of the BOADICEA multifactorial breast cancer risk prediction model in a large prospective cohort study. J Med Genet 2022; 59:1196-1205. [PMID: 36162852 PMCID: PMC9691822 DOI: 10.1136/jmg-2022-108806] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/24/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND The multifactorial Breast and Ovarian Analysis of Disease Incidence and Carrier Estimation Algorithm (BOADICEA) breast cancer risk prediction model has been recently extended to consider all established breast cancer risk factors. We assessed the clinical validity of the model in a large independent prospective cohort. METHODS We validated BOADICEA (V.6) in the Swedish KARolinska Mammography Project for Risk Prediction of Breast Cancer (KARMA) cohort including 66 415 women of European ancestry (median age 54 years, IQR 45-63; 816 incident breast cancers) without previous cancer diagnosis. We calculated 5-year risks on the basis of questionnaire-based risk factors, pedigree-structured first-degree family history, mammographic density (BI-RADS), a validated breast cancer polygenic risk score (PRS) based on 313-SNPs, and pathogenic variant status in 8 breast cancer susceptibility genes: BRCA1, BRCA2, PALB2, CHEK2, ATM, RAD51C, RAD51D and BARD1. Calibration was assessed by comparing observed and expected risks in deciles of predicted risk and the calibration slope. The discriminatory ability was assessed using the area under the curve (AUC). RESULTS Among the individual model components, the PRS contributed most to breast cancer risk stratification. BOADICEA was well calibrated in predicting the risks for low-risk and high-risk women when all, or subsets of risk factors are included in the risk prediction. Discrimination was maximised when all risk factors are considered (AUC=0.70, 95% CI: 0.66 to 0.73; expected-to-observed ratio=0.88, 95% CI: 0.75 to 1.04; calibration slope=0.97, 95% CI: 0.95 to 0.99). The full multifactorial model classified 3.6% women as high risk (5-year risk ≥3%) and 11.1% as very low risk (5-year risk <0.33%). CONCLUSION The multifactorial BOADICEA model provides valid breast cancer risk predictions and a basis for personalised decision-making on disease prevention and screening.
Collapse
Affiliation(s)
- Xin Yang
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Mikael Eriksson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Andrew Lee
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Goska Leslie
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Michael Lush
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Jean Wang
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Joe Dennis
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Leila Dorling
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Sara Carvalho
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Nasim Mavaddat
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| | - Jacques Simard
- Department of Molecular Medicine, Université Laval and CHU de Québec-Université Laval Research Center, Quebec City, Quebec, Canada
| | - Marjanka K Schmidt
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
- Devision of Molecular Pathology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Södersjukhuset, Stockholm, Sweden
| | - Antonis C Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, UK
| |
Collapse
|
122
|
Taylor G, McWilliams L, Woof VG, Evans DG, French DP. What are the views of three key stakeholder groups on extending the breast screening interval for low-risk women? A secondary qualitative analysis. Health Expect 2022; 25:3287-3296. [PMID: 36305519 PMCID: PMC9700144 DOI: 10.1111/hex.13637] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/14/2022] [Accepted: 10/16/2022] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION There is increasing interest in risk-stratified breast screening, whereby the prevention and early detection offers vary by a woman's estimated risk of breast cancer. To date, more focus has been directed towards high-risk screening pathways rather than considering women at lower risk, who may be eligible for extended screening intervals. This secondary data analysis aimed to compare the views of three key stakeholder groups on how extending screening intervals for low-risk women should be implemented and communicated as part of a national breast screening programme. METHODS Secondary data analysis of three qualitative studies exploring the views of distinct stakeholder groups was conducted. Interviews took place with 23 low-risk women (identified from the BC-Predict study) and 17 national screening figures, who were involved in policy-making and implementation. In addition, three focus groups and two interviews were conducted with 26 healthcare professionals. A multiperspective thematic analysis was conducted to identify similarities and differences between stakeholders. FINDINGS Three themes were produced: Questionable assumptions about negative consequences, highlighting how other stakeholders lack trust in how women are likely to understand extended screening intervals; Preserving the integrity of the programme, centring on decision-making and maintaining a positive reputation of breast screening and Negotiating a communication pathway highlighting communication expectations and public campaign importance. CONCLUSIONS A risk-stratified screening programme should consider how best to engage women assessed as having a low risk of breast cancer to ensure mutual trust, balance the practicality of change whilst ensuring acceptability, and carefully develop multilevel inclusive communication strategies. PATIENT AND PUBLIC CONTRIBUTION The research within this paper involved patient/public contributors throughout including study design and materials input.
Collapse
Affiliation(s)
- Grace Taylor
- School of Health Sciences, Manchester Centre of Health Psychology, Division of Psychology and Mental HealthUniversity of ManchesterManchesterUK
| | - Lorna McWilliams
- School of Health Sciences, Manchester Centre of Health Psychology, Division of Psychology and Mental HealthUniversity of ManchesterManchesterUK
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science CentreCentral Manchester University Hospitals NHS Foundation TrustManchesterUK
| | - Victoria G. Woof
- School of Health Sciences, Manchester Centre of Health Psychology, Division of Psychology and Mental HealthUniversity of ManchesterManchesterUK
| | - D. Gareth Evans
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science CentreCentral Manchester University Hospitals NHS Foundation TrustManchesterUK
- The Nightingale and Prevent Breast Cancer CentreManchester University NHS Foundation TrustManchesterUK
- Manchester Breast Centre, Manchester Cancer Research CentreUniversity of ManchesterManchesterUK
- Genomic Medicine, Division of Evolution and Genomic Sciences, St Mary's Hospital, Manchester University NHS Foundation TrustThe University of ManchesterManchesterUK
| | - David P. French
- School of Health Sciences, Manchester Centre of Health Psychology, Division of Psychology and Mental HealthUniversity of ManchesterManchesterUK
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science CentreCentral Manchester University Hospitals NHS Foundation TrustManchesterUK
- Manchester Breast Centre, Manchester Cancer Research CentreUniversity of ManchesterManchesterUK
| |
Collapse
|
123
|
Solary E, Blanc P, Boutros M, Girvalaki C, Locatelli F, Medema RH, Nagy P, Tabernero J. UNCAN.eu, a European Initiative to UNderstand CANcer. Cancer Discov 2022; 12:2504-2508. [PMID: 36074491 DOI: 10.1158/2159-8290.cd-22-0970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
"UNCAN.eu" refers to a collective European effort seeking to enable a leap forward in our understanding of cancer. This initiative, which includes the creation of a European cancer research data hub, will pave the way to new advances in cancer care. Starting on September 1, 2022, a 15-month coordination and support action will generate a blueprint for UNCAN.eu. Here, we summarize the cancer research issues that the blueprint will propose to tackle at the European level.
Collapse
Affiliation(s)
- Eric Solary
- Université Paris-Saclay and INSERM, Gustave Roussy Cancer Center, Villejuif, France
| | | | - Michael Boutros
- German Cancer Research Center (DKFZ) and Heidelberg University, Heidelberg, Germany
| | | | - Franco Locatelli
- Bambino Gesù Children's Hospital, University of the Sacred Heart, Rome, Italy
| | - Rene H Medema
- Oncode Institute and The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Péter Nagy
- National Institute of Oncology and National Tumor Biology Laboratory, Budapest, Hungary
| | - Josep Tabernero
- Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
124
|
Hughes E, Wagner S, Pruss D, Bernhisel R, Probst B, Abkevich V, Simmons T, Hullinger B, Judkins T, Rosenthal E, Roa B, Domchek SM, Eng C, Garber J, Gary M, Klemp J, Mukherjee S, Offit K, Olopade OI, Vijai J, Weitzel JN, Whitworth P, Yehia L, Gordon O, Pederson H, Kurian A, Slavin TP, Gutin A, Lanchbury JS. Development and Validation of a Breast Cancer Polygenic Risk Score on the Basis of Genetic Ancestry Composition. JCO Precis Oncol 2022; 6:e2200084. [PMID: 36331239 PMCID: PMC9666117 DOI: 10.1200/po.22.00084] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 07/11/2022] [Accepted: 09/08/2022] [Indexed: 08/12/2023] Open
Abstract
PURPOSE Polygenic risk scores (PRSs) for breast cancer (BC) risk stratification have been developed primarily in women of European ancestry. Their application to women of non-European ancestry has lagged because of the lack of a formal approach to incorporate genetic ancestry and ancestry-dependent variant frequencies and effect sizes. Here, we propose a multiple-ancestry PRS (MA-PRS) that addresses these issues and may be useful in the development of equitable PRSs across other cancers and common diseases. MATERIALS AND METHODS Women referred for hereditary cancer testing were divided into consecutive cohorts for development (n = 189,230) and for independent validation (n = 89,126). Individual genetic composition as fractions of three reference ancestries (African, East Asian, and European) was determined from ancestry-informative single-nucleotide polymorphisms. The MA-PRS is a combination of three ancestry-specific PRSs on the basis of genetic ancestral composition. Stratification of risk was evaluated by multivariable logistic regression models controlling for family cancer history. Goodness-of-fit analysis compared expected with observed relative risks by quantiles of the MA-PRS distribution. RESULTS In independent validation, the MA-PRS was significantly associated with BC risk in the full cohort (odds ratio, 1.43; 95% CI, 1.40 to 1.46; P = 8.6 × 10-308) and within each major ancestry. The top decile of the MA-PRS consistently identified patients with two-fold increased risk of developing BC. Goodness-of-fit tests showed that the MA-PRS was well calibrated and predicted BC risk accurately in the tails of the distribution for both European and non-European women. CONCLUSION The MA-PRS uses genetic ancestral composition to expand the utility of polygenic risk prediction to non-European women. Inclusion of genetic ancestry in polygenic risk prediction presents an opportunity for more personalized treatment decisions for women of varying and mixed ancestries.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Susan M. Domchek
- Basser Center for BRCA, University of Pennsylvania, Philadelphia, PA
| | - Charis Eng
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH
| | | | | | - Jennifer Klemp
- The University of Kansas Cancer Center, The University of Kansas Medical Center, Kansas City, KS
| | | | - Kenneth Offit
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Joseph Vijai
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | - Lamis Yehia
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Ora Gordon
- Providence Health and Services, Renton, WA
| | - Holly Pederson
- Medical Breast Services, Cleveland Clinic, Cleveland, OH
| | | | | | | | | |
Collapse
|
125
|
Hawkins R, McWilliams L, Ulph F, Evans DG, French DP. Healthcare professionals' views following implementation of risk stratification into a national breast cancer screening programme. BMC Cancer 2022; 22:1058. [PMID: 36224549 PMCID: PMC9555254 DOI: 10.1186/s12885-022-10134-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Background It is crucial to determine feasibility of risk-stratified screening to facilitate successful implementation. We introduced risk-stratification (BC-Predict) into the NHS Breast Screening Programme (NHSBSP) at three screening sites in north-west England from 2019 to 2021. The present study investigated the views of healthcare professionals (HCPs) on acceptability, barriers, and facilitators of the BC-Predict intervention and on the wider implementation of risk-based screening after BC-Predict was implemented in their screening site. Methods Fourteen semi-structured interviews were conducted with HCPs working across the breast screening pathway at three NHSBSP sites that implemented BC-Predict. Thematic analysis interpreted the data. Results Three pre-decided themes were produced. (1) Acceptability of risk-based screening: risk-stratification was perceived as a beneficial step for both services and women. HCPs across the pathway reported low burden of running the BC-Predict trial on routine tasks, but with some residual concerns; (2) Barriers to implementation: comprised capacity constraints of services including the inadequacy of current IT systems to manage women with different risk profiles and, (3) Facilitators to implementation: included the continuation of stakeholder consultation across the pathway to inform implementation and need for dedicated risk screening admin staff, a push for mammography staff recruitment and guidance for screening services. Telephone helplines, integrating primary care, and supporting access for all language needs was emphasised. Conclusion Risk-stratified breast screening was viewed as a progressive step providing it does not worsen inequalities for women. Implementation of risk-stratified breast screening requires staff to be reassured that there will be systems in place to support implementation and that it will not further burden their workload. Next steps require a comprehensive assessment of the resource needed for risk-stratification versus current resource availability, upgrades to screening IT and building screening infrastructure. The role of primary care needs to be determined. Simplification and clarification of risk-based screening pathways is needed to support HCPs agency and facilitate implementation. Forthcoming evidence from ongoing randomised controlled trials assessing effectiveness of breast cancer risk-stratification will also determine implementation. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10134-0.
Collapse
Affiliation(s)
- Rachel Hawkins
- The Christie NHS Foundation Trust, Wilmslow Rd, Manchester, M20 4BX, UK. .,NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, England.
| | - Lorna McWilliams
- Manchester Centre for Health Psychology, Division of Psychology & Mental Health, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, England
| | - Fiona Ulph
- Manchester Centre for Health Psychology, Division of Psychology & Mental Health, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - D Gareth Evans
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, England.,Nightingale & Prevent Breast Cancer Research Unit, Manchester University NHS Foundation Trust, Southmoor Road, M23 9LT, Wythenshawe, Manchester, UK.,Department of Genomic Medicine, Division of Evolution and Genomic Science, Manchester Academic Health Science Centre, University of Manchester, Manchester University NHS Foundation Trust, Oxford Road, M13 9WL, Manchester, UK
| | - David P French
- Manchester Centre for Health Psychology, Division of Psychology & Mental Health, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.,NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, England
| |
Collapse
|
126
|
Nickel B, Copp T, Li T, Dolan H, Brennan M, Verde A, Vaccaro L, McCaffery K, Houssami N. A systematic assessment of online international breast density information. Breast 2022; 65:23-31. [PMID: 35763979 PMCID: PMC9240362 DOI: 10.1016/j.breast.2022.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Breast density has become a topic of international discussion due to its associated risk of breast cancer. As online is often a primary source of women's health information it is therefore essential that breast density information it is understandable, accurate and reflects the best available evidence. This study aimed to systematically assess online international breast density information including recommendations to women. METHODS Searches were conducted from five different English-speaking country-specific Google locations. Relevant breast density information was extracted from the identified websites. Readability was assessed using the SHeLL Editor, and understandability and actionability using the Patient Education Materials Assessment Tool (PEMAT). A content analysis of specific recommendations to women was also conducted. RESULTS Forty-two eligible websites were identified and systematically assessed. The included informational content varied across websites. The average grade reading level across all websites was 12.4 (range 8.9-15.4). The mean understandability was 69.9% and the mean actionability was 40.1%, with 18/42 and 39/42 websites respectively scoring lower than adequate (70%). Thirty-six (85.7%) of the websites had breast density-related recommendation to women, with 'talk to your doctor' (n = 33, 78.6%) the most common. CONCLUSIONS Online information about breast density varies widely and is not generally presented in a way that women can easily understand and act on, therefore greatly reducing the ability for informed decision-making. International organisations and groups disseminating breast density information need to ensure that women are presented with health literacy-sensitive and balanced information, and be aware of the impact that recommendations may have on practice.
Collapse
Affiliation(s)
- Brooke Nickel
- Wiser Healthcare, Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Sydney Health Literacy Lab, Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.
| | - Tessa Copp
- Wiser Healthcare, Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Sydney Health Literacy Lab, Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Tong Li
- The Daffodil Centre, The University of Sydney, A Joint Venture with Cancer Council NSW, Sydney, Australia
| | - Hankiz Dolan
- Wiser Healthcare, Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Meagan Brennan
- The University of Notre Dame Australia, School of Medicine Sydney, Sydney, Australia; Westmead Breast Cancer Institute, Westmead Hospital, Sydney, Australia
| | - Angela Verde
- Breast Cancer Network Australia, Melbourne, Australia
| | - Lisa Vaccaro
- Health Consumers New South Wales, Sydney, Australia; Discipline of Behavioural and Social Sciences in Health, Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Kirsten McCaffery
- Wiser Healthcare, Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; Sydney Health Literacy Lab, Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Nehmat Houssami
- Wiser Healthcare, Sydney School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia; The Daffodil Centre, The University of Sydney, A Joint Venture with Cancer Council NSW, Sydney, Australia
| |
Collapse
|
127
|
Marzo-Castillejo M, Bartolomé-Moreno C, Bellas-Beceiro B, Melús-Palazón E, Vela-Vallespín C. [PAPPS Expert Groups. Cancer prevention recommendations: Update 2022]. Aten Primaria 2022; 54 Suppl 1:102440. [PMID: 36435580 PMCID: PMC9705215 DOI: 10.1016/j.aprim.2022.102440] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer is a major cause of morbidity and mortality. Tobacco use, unhealthy diet, and physical inactivity are some of the lifestyle risk factors that have led to an increase in cancer. This article updates the evidence and includes recommendations for prevention strategies for each of the cancers with the highest incidence. These are based on the reduction of risk factors (primary prevention) and early diagnosis of cancer through screening and early detection of signs and symptoms, in medium-risk and high-risk populations. This update of the 2022 PAPPS has taken into account the vision of the National Health System Cancer Strategy, an update approved by the Interterritorial Council of the National Health System on January 2021 and the European Strategy (Europe's Beating Cancer Plan) presented on 4 February 2021.
Collapse
Affiliation(s)
- Mercè Marzo-Castillejo
- Unitat de Suport a la Recerca Metropolitana Sud, IDIAP Jordi Gol, Direcció d'Atenció Primària Costa de Ponent, Institut Català de la Salut, Barcelona, España.
| | - Cruz Bartolomé-Moreno
- Centro de Salud Parque Goya de Zaragoza y Unidad Docente de Atención Familiar y Comunitaria Sector Zaragoza I, Servicio Aragonés de Salud, Zaragoza, España
| | - Begoña Bellas-Beceiro
- Unidad Docente de Atención Familiar y Comunitaria La Laguna-Tenerife Norte, Complejo Hospitalario Universitario de Canarias, La Laguna, Santa Cruz de Tenerife, España
| | - Elena Melús-Palazón
- Centro de Salud Actur Oeste de Zaragoza y Unidad Docente de Atención Familiar y Comunitaria Sector Zaragoza I, Servicio Aragonés de Salud, Zaragoza, España
| | - Carmen Vela-Vallespín
- ABS del Riu Nord i Riu Sud, Institut Català de la Salut, Santa Coloma de Gramenet, Barcelona, España
| |
Collapse
|
128
|
Gastounioti A, Eriksson M, Cohen EA, Mankowski W, Pantalone L, Ehsan S, McCarthy AM, Kontos D, Hall P, Conant EF. External Validation of a Mammography-Derived AI-Based Risk Model in a U.S. Breast Cancer Screening Cohort of White and Black Women. Cancers (Basel) 2022; 14:4803. [PMID: 36230723 PMCID: PMC9564051 DOI: 10.3390/cancers14194803] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Despite the demonstrated potential of artificial intelligence (AI) in breast cancer risk assessment for personalizing screening recommendations, further validation is required regarding AI model bias and generalizability. We performed external validation on a U.S. screening cohort of a mammography-derived AI breast cancer risk model originally developed for European screening cohorts. We retrospectively identified 176 breast cancers with exams 3 months to 2 years prior to cancer diagnosis and a random sample of 4963 controls from women with at least one-year negative follow-up. A risk score for each woman was calculated via the AI risk model. Age-adjusted areas under the ROC curves (AUCs) were estimated for the entire cohort and separately for White and Black women. The Gail 5-year risk model was also evaluated for comparison. The overall AUC was 0.68 (95% CIs 0.64−0.72) for all women, 0.67 (0.61−0.72) for White women, and 0.70 (0.65−0.76) for Black women. The AI risk model significantly outperformed the Gail risk model for all women p < 0.01 and for Black women p < 0.01, but not for White women p = 0.38. The performance of the mammography-derived AI risk model was comparable to previously reported European validation results; non-significantly different when comparing White and Black women; and overall, significantly higher than that of the Gail model.
Collapse
Affiliation(s)
- Aimilia Gastounioti
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mikael Eriksson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Eric A. Cohen
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Walter Mankowski
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lauren Pantalone
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah Ehsan
- Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anne Marie McCarthy
- Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Despina Kontos
- Center for Biomedical Image Computing and Analytics (CBICA), University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Oncology, Södersjukhuset, 118 83 Stockholm, Sweden
| | - Emily F. Conant
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
129
|
McWilliams L, Evans DG, Payne K, Harrison F, Howell A, Howell SJ, French DP. Implementing Risk-Stratified Breast Screening in England: An Agenda Setting Meeting. Cancers (Basel) 2022; 14:cancers14194636. [PMID: 36230559 PMCID: PMC9563640 DOI: 10.3390/cancers14194636] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
It is now possible to accurately assess breast cancer risk at routine NHS Breast Screening Programme (NHSBSP) appointments, provide risk feedback and offer risk management strategies to women at higher risk. These strategies include National Institute for Health and Care Excellence (NICE) approved additional breast screening and risk-reducing medication. However, the NHSBSP invites nearly all women three-yearly, regardless of risk. In March 2022, a one-day agenda setting meeting took place in Manchester to discuss the feasibility and desirability of implementation of risk-stratified screening in the NHSBSP. Fifty-eight individuals participated (38 face-to-face, 20 virtual) with relevant expertise from academic, clinical and/or policy-making perspectives. Key findings were presented from the PROCAS2 NIHR programme grant regarding feasibility of risk-stratified screening in the NHSBSP. Participants discussed key uncertainties in seven groups, followed by a plenary session. Discussions were audio-recorded and thematically analysed to produce descriptive themes. Five themes were developed: (i) risk and health economic modelling; (ii) health inequalities and communication with women; (iii); extending screening intervals for low-risk women; (iv) integration with existing NHSBSP; and (v) potential new service models. Most attendees expected some form of risk-stratified breast screening to be implemented in England and collectively identified key issues to be resolved to facilitate this.
Collapse
Affiliation(s)
- Lorna McWilliams
- Manchester Centre for Health Psychology, Division of Psychology & Mental Health, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PL, UK
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester M13 9WU, UK
- Correspondence:
| | - D. Gareth Evans
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester M13 9WU, UK
- Genomic Medicine, Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, St Mary’s Hospital, Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK
- Nightingale & Prevent Breast Cancer Research Unit, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester M23 9LT, UK
- Manchester Breast Centre, Manchester Cancer Research Centre, University of Manchester, 55 Wilmslow Road, Manchester M20 4GJ, UK
| | - Katherine Payne
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester M13 9WU, UK
- Manchester Centre for Health Economics, School of Health Sciences, Faculty of Biology Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | | | - Anthony Howell
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester M13 9WU, UK
- Nightingale & Prevent Breast Cancer Research Unit, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester M23 9LT, UK
- Manchester Breast Centre, Manchester Cancer Research Centre, University of Manchester, 55 Wilmslow Road, Manchester M20 4GJ, UK
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Sacha J. Howell
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester M13 9WU, UK
- Nightingale & Prevent Breast Cancer Research Unit, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Southmoor Road, Wythenshawe, Manchester M23 9LT, UK
- Manchester Breast Centre, Manchester Cancer Research Centre, University of Manchester, 55 Wilmslow Road, Manchester M20 4GJ, UK
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - David P. French
- Manchester Centre for Health Psychology, Division of Psychology & Mental Health, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PL, UK
- NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester M13 9WU, UK
- Manchester Breast Centre, Manchester Cancer Research Centre, University of Manchester, 55 Wilmslow Road, Manchester M20 4GJ, UK
| | | |
Collapse
|
130
|
[CD40LG is a novel immune- and stroma-related prognostic biomarker in the tumor microenvironment of invasive breast cancer]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:1267-1278. [PMID: 36210698 PMCID: PMC9550551 DOI: 10.12122/j.issn.1673-4254.2022.09.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE To identify tumor microenvironment (TME)- related genes associated with the occurrence of invasive breast cancer as potential prognostic biomarkers and therapeutic targets. METHODS RNA transcriptome data and clinically relevant data were retrieved from TCGA database, and the StromalScore and ImmuneScore were calculated using the ESTIMATE algorithm. The differentially expressed genes (DEGs) were screened by taking the intersection. A protein- protein interaction network was established, and univariate COX regression analysis was used to identify the core genes among the DEGs. A core gene was selected for GSEA and CIBERSORT analysis to determine the function of the core gene and the proportion of tumor-infiltrating immune cells, respectively. Western blotting and qRT-PCR were performed to verify the expression level of CD40LG in breast cancer cell lines and clinical specimens. RESULTS A total of 1222 samples (124 normal and 1098 tumor samples) were extracted from TCGA for analysis, from which 487 DEGs were identified. These genes were mainly enriched in immune-related pathways, and crossover analysis identified 11 key genes (CD40LG, ITK, CD5, CD3E, SPN, IL7R, CD48, CCL19, CD2, CD52, and CD2711) associated with breast cancer TME status. CD40LG was selected as the core gene, whose high expression was found to be associated with a longer overall survival of breast cancer patients (P=0.002), and its expression level differed significantly with TNM stage and tumor size (P < 0.05). GSEA and CIBERSORT analyses indicated that CD40LG expression level was associated with immune activity in the TME. Western blotting and qRT-PCR showed that the protein and mRNA expression of CD40LG were significantly lower in breast cancer cells and cancer tissues than in normal breast cells and adjacent tissues. CONCLUSIONS The high expression of CD40LG in TME is positively correlated with the survival of patients with invasive breast cancer, suggesting its value as a potential new biomarker for predicting prognosis of the patients.
Collapse
|
131
|
Hung RJ, Khodayari Moez E, Kim SJ, Budhathoki S, Brooks JD. Considerations of biomarker application for cancer continuum in the era of precision medicine. CURR EPIDEMIOL REP 2022; 9:200-211. [PMID: 36090700 PMCID: PMC9454320 DOI: 10.1007/s40471-022-00295-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2022] [Indexed: 11/25/2022]
Abstract
Purpose of the review The goal of this review is to highlight emerging biomarker research by the key phases of the cancer continuum and outline the methodological considerations for biomarker application. Recent findings While biomarkers have an established role in targeted therapy and to some extent, disease monitoring, their role in early detection and survivorship remains to be elucidated. With the advent of omics technology, the discovery of biomarkers has been accelerated exponentially, therefore careful consideration to ensure an unbiased study design and robust validity is crucial. Summary The rigor of biomarker research holds the key to the success of precision health care. The potential clinical utility and the feasibility of implementation should be central to future biomarker research study design.
Collapse
Affiliation(s)
- Rayjean J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Elham Khodayari Moez
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada
| | - Shana J Kim
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Sanjeev Budhathoki
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada
| | - Jennifer D Brooks
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| |
Collapse
|
132
|
Laza-Vásquez C, Martínez-Alonso M, Forné-Izquierdo C, Vilaplana-Mayoral J, Cruz-Esteve I, Sánchez-López I, Reñé-Reñé M, Cazorla-Sánchez C, Hernández-Andreu M, Galindo-Ortego G, Llorens-Gabandé M, Pons-Rodríguez A, Rué M. Feasibility and Acceptability of Personalized Breast Cancer Screening (DECIDO Study): A Single-Arm Proof-of-Concept Trial. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:10426. [PMID: 36012059 PMCID: PMC9407798 DOI: 10.3390/ijerph191610426] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/11/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
The aim of this study was to assess the acceptability and feasibility of offering risk-based breast cancer screening and its integration into regular clinical practice. A single-arm proof-of-concept trial was conducted with a sample of 387 women aged 40-50 years residing in the city of Lleida (Spain). The study intervention consisted of breast cancer risk estimation, risk communication and screening recommendations, and a follow-up. A polygenic risk score with 83 single nucleotide polymorphisms was used to update the Breast Cancer Surveillance Consortium risk model and estimate the 5-year absolute risk of breast cancer. The women expressed a positive attitude towards varying the frequency of breast screening according to individual risk and, especially, more frequently inviting women at higher-than-average risk. A lower intensity screening for women at lower risk was not as welcome, although half of the participants would accept it. Knowledge of the benefits and harms of breast screening was low, especially with regard to false positives and overdiagnosis. The women expressed a high understanding of individual risk and screening recommendations. The participants' intention to participate in risk-based screening and satisfaction at 1-year were very high.
Collapse
Affiliation(s)
- Celmira Laza-Vásquez
- Department of Nursing and Physiotherapy and Health Care Research Group (GRECS), IRBLleida—Institut de Recerca Biomèdica de Lleida, University of Lleida, 25198 Lleida, Spain
| | - Montserrat Martínez-Alonso
- IRBLleida—Institut de Recerca Biomèdica de Lleida, Department of Basic Medical Sciences, University of Lleida, 25198 Lleida, Spain
| | - Carles Forné-Izquierdo
- Department of Basic Medical Sciences, University of Lleida, 25198 Lleida, Spain
- Heorfy Consulting, 25007 Lleida, Spain
| | - Jordi Vilaplana-Mayoral
- Department of Computing and Industrial Engineering, University of Lleida, 25001 Lleida, Spain
| | - Inés Cruz-Esteve
- Primer de Maig Basic Health Area, Catalan Institute of Health, 25003 Lleida, Spain
| | | | - Mercè Reñé-Reñé
- Department of Radiology, Arnau de Vilanova University Hospital, 25198 Lleida, Spain
| | | | | | | | | | - Anna Pons-Rodríguez
- Example Basic Health Area, Catalan Institute of Health, 25006 Lleida, Spain
- Health PhD Program, University of Lleida, 25198 Lleida, Spain
| | - Montserrat Rué
- IRBLleida—Institut de Recerca Biomèdica de Lleida, Department of Basic Medical Sciences, University of Lleida, 25198 Lleida, Spain
| |
Collapse
|
133
|
Wang Y, Tsuo K, Kanai M, Neale BM, Martin AR. Challenges and Opportunities for Developing More Generalizable Polygenic Risk Scores. Annu Rev Biomed Data Sci 2022; 5:293-320. [PMID: 35576555 PMCID: PMC9828290 DOI: 10.1146/annurev-biodatasci-111721-074830] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Polygenic risk scores (PRS) estimate an individual's genetic likelihood of complex traits and diseases by aggregating information across multiple genetic variants identified from genome-wide association studies. PRS can predict a broad spectrum of diseases and have therefore been widely used in research settings. Some work has investigated their potential applications as biomarkers in preventative medicine, but significant work is still needed to definitively establish and communicate absolute risk to patients for genetic and modifiable risk factors across demographic groups. However, the biggest limitation of PRS currently is that they show poor generalizability across diverse ancestries and cohorts. Major efforts are underway through methodological development and data generation initiatives to improve their generalizability. This review aims to comprehensively discuss current progress on the development of PRS, the factors that affect their generalizability, and promising areas for improving their accuracy, portability, and implementation.
Collapse
Affiliation(s)
- Ying Wang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA;
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Kristin Tsuo
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA;
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Masahiro Kanai
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA;
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, USA
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Benjamin M Neale
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA;
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Alicia R Martin
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, Massachusetts, USA;
- Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| |
Collapse
|
134
|
French DP, McWilliams L, Howell A, Evans DG. Does receiving high or low breast cancer risk estimates produce a reduction in subsequent breast cancer screening attendance? Cohort study. Breast 2022; 64:47-49. [PMID: 35569186 PMCID: PMC9111984 DOI: 10.1016/j.breast.2022.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/27/2022] [Accepted: 05/05/2022] [Indexed: 11/01/2022] Open
Abstract
Risk-stratified breast cancer screening may improve the balance of screening benefits to harms. We assess a potential new harm: reduced screening attendance in women receiving below average-risk (false reassurance) or higher-risk results (screening avoidance). Following initial screening, 26,668 women in the PROCAS study received breast cancer risk estimates, with attendance recorded for two subsequent screening rounds. First-screen attendance was slightly reduced in below-average (85.6%) but not higher-risk women, compared to other women (86.4%). Second-screen attendance increased for women at higher-risk (89.2%) but not below-average, compared to other women (78.8%). Concerns about this potential harm of risk-stratified screening therefore appear unfounded. A potential harm of risk-stratified screening is a reduction in future attendance. Some evidence of a small reduction in future attendance in below-average women. Large increase in attendance in higher-risk women at future screening rounds.
Collapse
|
135
|
Nabi H. Personalized Approaches for the Prevention and Treatment of Breast Cancer. J Pers Med 2022; 12:jpm12081201. [PMID: 35893295 PMCID: PMC9331702 DOI: 10.3390/jpm12081201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
Breast cancer (BC) remains a major public health issue worldwide [...]
Collapse
Affiliation(s)
- Hermann Nabi
- Oncology Division, CHU de Québec-Université Laval Research Center, Quebec City, QC G1S 4L8, Canada; ; Tel.: +1-418-682-7511 (ext. 82800)
- Université Laval Cancer Research Center (CRC), Université Laval, Quebec City, QC G1S 4L8, Canada
- Department of Social and Preventive Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1S 4L8, Canada
| |
Collapse
|
136
|
Molecular perspective on targeted therapy in breast cancer: a review of current status. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:149. [PMID: 35834030 PMCID: PMC9281252 DOI: 10.1007/s12032-022-01749-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/11/2022] [Indexed: 12/24/2022]
Abstract
Breast cancer is categorized at the molecular level according to the status of certain hormone and growth factor receptors, and this classification forms the basis of current diagnosis and treatment. The development of resistance to treatment and recurrence of the disease have led researchers to develop new therapies. In recent years, most of the research in the field of oncology has focused on the development of targeted therapies, which are treatment methods developed directly against molecular abnormalities. Promising advances have been made in clinical trials investigating the effect of these new treatment modalities and their combinations with existing therapeutic treatments in the treatment of breast cancer. Monoclonal antibodies, tyrosine kinase inhibitors, antibody–drug conjugates, PI3K/Akt/mTOR pathway inhibitors, cyclin-dependent kinase 4/6 inhibitors, anti-angiogenic drugs, PARP inhibitors are among the targeted therapies used in breast cancer treatment. In this review, we aim to present a molecular view of recently approved target agents used in breast cancer.
Collapse
|
137
|
Koopaie M, Kolahdooz S, Fatahzadeh M, Manifar S. Salivary biomarkers in breast cancer diagnosis: A systematic review and diagnostic meta-analysis. Cancer Med 2022; 11:2644-2661. [PMID: 35315584 PMCID: PMC9249990 DOI: 10.1002/cam4.4640] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/25/2021] [Accepted: 01/02/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Salivary diagnostics and their utility as a nonaggressive approach for breast cancer diagnosis have been extensively studied in recent years. This meta-analysis assesses the diagnostic value of salivary biomarkers in differentiating between patients with breast cancer and controls. METHODS We conducted a meta-analysis and systematic review of studies related to salivary diagnostics published in PubMed, EMBASE, Scopus, Ovid, Science Direct, Web of Science (WOS), and Google Scholar. The articles were chosen utilizing inclusion and exclusion criteria, as well as assessing their quality. Specificity and sensitivity, along with negative and positive likelihood ratios (NLR and PLR) and diagnostic odds ratio (DOR), were calculated based on random- or fixed-effects model. Area under the curve (AUC) and summary receiver-operating characteristic (SROC) were plotted and evaluated, and Fagan's Nomogram was evaluated for clinical utility. RESULTS Our systematic review and meta-analysis included 14 papers containing 121 study units with 8639 adult subjects (4149 breast cancer patients and 4490 controls without cancer). The pooled specificity and sensitivity were 0.727 (95% CI: 0.713-0.740) and 0.717 (95% CI: 0.703-0.730), respectively. The pooled NLR and PLR were 0.396 (95% CI: 0.364-0.432) and 2.597 (95% CI: 2.389-2.824), respectively. The pooled DOR was 7.837 (95% CI: 6.624-9.277), with the AUC equal to 0.801. The Fagan's nomogram showed post-test probabilities of 28% and 72% for negative and positive outcomes, respectively. We also conducted subgroup analyses to determine specificity, sensitivity, DOR, PLR, and NLR based on the mean age of patients (≤52 or >52 years old), saliva type (stimulated and unstimulated saliva), biomarker measurement method (mass spectrometry [MS] and non-MS measurement methods), sample size (≤55 or >55), biomarker type (proteomics, metabolomics, transcriptomics and proteomics, and reagent-free biophotonic), and nations. CONCLUSION Saliva, as a noninvasive biomarker, has the potential to accurately differentiate breast cancer patients from healthy controls.
Collapse
Affiliation(s)
| | | | - Mahnaz Fatahzadeh
- Department of Diagnostic SciencesRutgers School of Dental MedicineNewarkNew JerseyUSA
| | - Soheila Manifar
- Tehran University of Medical SciencesTehranIran
- Cancer Research Center, Cancer Institute of IranTehranIran
| |
Collapse
|
138
|
Thomassin-Naggara I, Ceugnart L, Tardivon A, Verzaux L, Balleyguier C, Taourel P, Seradour B. Intelligence artificielle : Place dans le dépistage du cancer du sein en France. Bull Cancer 2022; 109:780-785. [DOI: 10.1016/j.bulcan.2022.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/24/2022] [Accepted: 04/11/2022] [Indexed: 01/20/2023]
|
139
|
Bartlett TE, Evans I, Jones A, Barrett JE, Haran S, Reisel D, Papaikonomou K, Jones L, Herzog C, Pashayan N, Simões BM, Clarke RB, Evans DG, Ghezelayagh TS, Ponandai-Srinivasan S, Boggavarapu NR, Lalitkumar PG, Howell SJ, Risques RA, Rådestad AF, Dubeau L, Gemzell-Danielsson K, Widschwendter M. Antiprogestins reduce epigenetic field cancerization in breast tissue of young healthy women. Genome Med 2022; 14:64. [PMID: 35701800 PMCID: PMC9199133 DOI: 10.1186/s13073-022-01063-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/17/2022] [Indexed: 02/08/2023] Open
Abstract
Background Breast cancer is a leading cause of death in premenopausal women. Progesterone drives expansion of luminal progenitor cells, leading to the development of poor-prognostic breast cancers. However, it is not known if antagonising progesterone can prevent breast cancers in humans. We suggest that targeting progesterone signalling could be a means of reducing features which are known to promote breast cancer formation.
Methods In healthy premenopausal women with and without a BRCA mutation we studied (i) estrogen and progesterone levels in saliva over an entire menstrual cycle (n = 20); (ii) cancer-free normal breast-tissue from a control population who had no family or personal history of breast cancer and equivalently from BRCA1/2 mutation carriers (n = 28); triple negative breast cancer (TNBC) biopsies and healthy breast tissue taken from sites surrounding the TNBC in the same individuals (n = 14); and biopsies of ER+ve/PR+ve stage T1–T2 cancers and healthy breast tissue taken from sites surrounding the cancer in the same individuals (n = 31); and (iii) DNA methylation and DNA mutations in normal breast tissue (before and after treatment) from clinical trials that assessed the potential preventative effects of vitamins and antiprogestins (mifepristone and ulipristal acetate; n = 44).
Results Daily levels of progesterone were higher throughout the menstrual cycle of BRCA1/2 mutation carriers, raising the prospect of targeting progesterone signalling as a means of cancer risk reduction in this population. Furthermore, breast field cancerization DNA methylation signatures reflective of (i) the mitotic age of normal breast epithelium and (ii) the proportion of luminal progenitor cells were increased in breast cancers, indicating that luminal progenitor cells with elevated replicative age are more prone to malignant transformation. The progesterone receptor antagonist mifepristone reduced both the mitotic age and the proportion of luminal progenitor cells in normal breast tissue of all control women and in 64% of BRCA1/2 mutation carriers. These findings were validated by an alternate progesterone receptor antagonist, ulipristal acetate, which yielded similar results. Importantly, mifepristone reduced both the TP53 mutation frequency as well as the number of TP53 mutations in mitotic-age-responders. Conclusions These data support the potential usage of antiprogestins for primary prevention of poor-prognostic breast cancers. Trial registration Clinical trial 1 Mifepristone treatment prior to insertion of a levonorgestrel releasing intrauterine system for improved bleeding control – a randomized controlled trial, clinicaltrialsregister.eu, 2009-009014-40; registered on 20 July 2009. Clinical trial 2 The effect of a progesterone receptor modulator on breast tissue in women with BRCA1 and 2 mutations, clinicaltrials.gov, NCT01898312; registered on 07 May 2013. Clinical trial 3 A pilot prevention study of the effects of the anti- progestin Ulipristal Acetate (UA) on surrogate markers of breast cancer risk, clinicaltrialsregister.eu, 2015-001587-19; registered on 15 July 2015. Supplementary Information The online version contains supplementary material available at 10.1186/s13073-022-01063-5.
Collapse
Affiliation(s)
- Thomas E Bartlett
- Department of Statistical Science, University College London, London, WC1E 7HB, UK
| | - Iona Evans
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, 74 Huntley Street, London, WC1E 6AU, UK
| | - Allison Jones
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, 74 Huntley Street, London, WC1E 6AU, UK
| | - James E Barrett
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, 74 Huntley Street, London, WC1E 6AU, UK.,European Translational Oncology Prevention and Screening (EUTOPS) Institute, Universität Innsbruck, 6060, Hall in Tirol, Austria.,Research Institute for Biomedical Aging Research, Universität Innsbruck, 6020, Innsbruck, Austria
| | - Shaun Haran
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, 74 Huntley Street, London, WC1E 6AU, UK
| | - Daniel Reisel
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, 74 Huntley Street, London, WC1E 6AU, UK
| | - Kiriaki Papaikonomou
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Louise Jones
- Centre for Tumour Biology Department, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Chiara Herzog
- European Translational Oncology Prevention and Screening (EUTOPS) Institute, Universität Innsbruck, 6060, Hall in Tirol, Austria.,Research Institute for Biomedical Aging Research, Universität Innsbruck, 6020, Innsbruck, Austria
| | - Nora Pashayan
- Department of Applied Health Research, University College London, 1-19 Torrington Place, London, WC1E 7HB, UK
| | - Bruno M Simões
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK, England
| | - Robert B Clarke
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK, England
| | - D Gareth Evans
- University of Manchester, St. Mary's Hospital, and University Hospital of South Manchester, Manchester, UK
| | - Talayeh S Ghezelayagh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA.,Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, 98195, USA
| | - Sakthivignesh Ponandai-Srinivasan
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Nageswara R Boggavarapu
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Parameswaran G Lalitkumar
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Sacha J Howell
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK, England.,Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Rosa Ana Risques
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Angelique Flöter Rådestad
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Louis Dubeau
- Department of Pathology, Keck School of Medicine, USC/Norris Comprehensive Cancer Centre, University of Southern California, Los Angeles, USA
| | - Kristina Gemzell-Danielsson
- Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Martin Widschwendter
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, 74 Huntley Street, London, WC1E 6AU, UK. .,European Translational Oncology Prevention and Screening (EUTOPS) Institute, Universität Innsbruck, 6060, Hall in Tirol, Austria. .,Research Institute for Biomedical Aging Research, Universität Innsbruck, 6020, Innsbruck, Austria. .,Department of Women's and Children's Health, Division of Obstetrics and Gynecology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
140
|
Moorthie S, Babb de Villiers C, Burton H, Kroese M, Antoniou AC, Bhattacharjee P, Garcia-Closas M, Hall P, Schmidt MK. Towards implementation of comprehensive breast cancer risk prediction tools in health care for personalised prevention. Prev Med 2022; 159:107075. [PMID: 35526672 DOI: 10.1016/j.ypmed.2022.107075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/05/2022] [Accepted: 05/02/2022] [Indexed: 12/24/2022]
Abstract
Advances in knowledge about breast cancer risk factors have led to the development of more comprehensive risk models. These integrate information on a variety of risk factors such as lifestyle, genetics, family history, and breast density. These risk models have the potential to deliver more personalised breast cancer prevention. This is through improving accuracy of risk estimates, enabling more effective targeting of preventive options and creating novel prevention pathways through enabling risk estimation in a wider variety of populations than currently possible. The systematic use of risk tools as part of population screening programmes is one such example. A clear understanding of how such tools can contribute to the goal of personalised prevention can aid in understanding and addressing barriers to implementation. In this paper we describe how emerging models, and their associated tools can contribute to the goal of personalised healthcare for breast cancer through health promotion, early disease detection (screening) and improved management of women at higher risk of disease. We outline how addressing specific challenges on the level of communication, evidence, evaluation, regulation, and acceptance, can facilitate implementation and uptake.
Collapse
Affiliation(s)
- Sowmiya Moorthie
- PHG Foundation, University of Cambridge, Cambridge, UK; Cambridge Public Health, University of Cambridge School of Clinical Medicine, Forvie Site, Cambridge Biomedical Campus, Cambridge CB2 0SR, United Kingdom.
| | | | - Hilary Burton
- PHG Foundation, University of Cambridge, Cambridge, UK
| | - Mark Kroese
- PHG Foundation, University of Cambridge, Cambridge, UK
| | - Antonis C Antoniou
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Proteeti Bhattacharjee
- Division of Molecular Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Montserrat Garcia-Closas
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health (NIH), Bethesda, USA
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden; Department of Oncology, Södersjukhuset, Stockholm, Sweden
| | - Marjanka K Schmidt
- Division of Molecular Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands; Department of Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
141
|
Overdetection of Breast Cancer. Curr Oncol 2022; 29:3894-3910. [PMID: 35735420 PMCID: PMC9222123 DOI: 10.3390/curroncol29060311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 11/25/2022] Open
Abstract
Overdetection (often referred to as overdiagnosis) of cancer is the detection of disease, such as through a screening program, that would otherwise remain occult through an individual’s life. In the context of screening, this could occur for cancers that were slow growing or indolent, or simply because an unscreened individual would have died from some other cause before the cancer had surfaced clinically. The main harm associated with overdetection is the subsequent overdiagnosis and overtreatment of disease. In this article, the phenomenon is reviewed, the methods of estimation of overdetection are discussed and reasons for variability in such estimates are given, with emphasis on an analysis using Canadian data. Microsimulation modeling is used to illustrate the expected time course of cancer detection that gives rise to overdetection. While overdetection exists, the actual amount is likely to be much lower than the estimate used by the Canadian Task Force on Preventive Health Care. Furthermore, the issue is of greater significance in older rather than younger women due to competing causes of death. The particular challenge associated with in situ breast cancer is considered and possible approaches to avoiding overtreatment are suggested.
Collapse
|
142
|
Lee SH, Moon WK. Glandular Tissue Component on Breast Ultrasound in Dense Breasts: A New Imaging Biomarker for Breast Cancer Risk. Korean J Radiol 2022; 23:574-580. [PMID: 35617993 PMCID: PMC9174505 DOI: 10.3348/kjr.2022.0099] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/04/2022] [Accepted: 04/10/2022] [Indexed: 11/15/2022] Open
Affiliation(s)
- Su Hyun Lee
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
| | - Woo Kyung Moon
- Department of Radiology, Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
143
|
Roux A, Cholerton R, Sicsic J, Moumjid N, French DP, Giorgi Rossi P, Balleyguier C, Guindy M, Gilbert FJ, Burrion JB, Castells X, Ritchie D, Keatley D, Baron C, Delaloge S, de Montgolfier S. Study protocol comparing the ethical, psychological and socio-economic impact of personalised breast cancer screening to that of standard screening in the "My Personal Breast Screening" (MyPeBS) randomised clinical trial. BMC Cancer 2022; 22:507. [PMID: 35524202 PMCID: PMC9073478 DOI: 10.1186/s12885-022-09484-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/02/2022] [Indexed: 12/11/2022] Open
Abstract
Background The MyPeBS study is an ongoing randomised controlled trial testing whether a risk-stratified breast cancer screening strategy is non-inferior, or eventually superior, to standard age-based screening at reducing incidence of stage 2 or more cancers. This large European Commission-funded initiative aims to include 85,000 women aged 40 to 70 years, without prior breast cancer and not previously identified at high risk in six countries (Belgium, France, Italy, Israel, Spain, UK). A specific work package within MyPeBS examines psychological, socio-economic and ethical aspects of this new screening strategy. It compares women’s reported data and outcomes in both trial arms on the following issues: general anxiety, cancer-related worry, understanding of breast cancer screening strategy and information-seeking behaviour, socio-demographic and economic characteristics, quality of life, risk perception, intention to change health-related behaviours, satisfaction with the trial. Methods At inclusion, 3-months, 1-year and 4-years, each woman participating in MyPeBS is asked to fill online questionnaires. Descriptive statistics, bivariate analyses, subgroup comparisons and analysis of variations over time will be performed with appropriate tests to assess differences between arms. Multivariate regression models will allow modelling of different patient reported data and outcomes such as comprehension of the information provided, general anxiety or cancer worry, and information seeking behaviour. In addition, a qualitative study (48 semi-structured interviews conducted in France and in the UK with women randomised in the risk-stratified arm), will help further understand participants’ acceptability and comprehension of the trial, and their experience of risk assessment. Discussion Beyond the scientific and medical objectives of this clinical study, it is critical to acknowledge the consequences of such a paradigm shift for women. Indeed, introducing a risk-based screening relying on individual biological differences also implies addressing non-biological differences (e.g. social status or health literacy) from an ethical perspective, to ensure equal access to healthcare. The results of the present study will facilitate making recommendations on implementation at the end of the trial to accompany any potential change in screening strategy. Trial registration Study sponsor: UNICANCER. My personalised breast screening (MyPeBS). Clinicaltrials.gov (2018) available at: https://clinicaltrials.gov/ct2/show/NCT03672331 Contact: Cécile VISSAC SABATIER, PhD, + 33 (0)1 73 79 77 58 ext + 330,142,114,293, contact@mypebs.eu. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09484-6.
Collapse
Affiliation(s)
- Alexandra Roux
- IRIS (UMR8156 CNRS & U997 INSERM), Paris 13 University, Aubervilliers, France
| | | | | | - Nora Moumjid
- Université Lyon 1, P2S EA 4129, Centre Léon Bérard, F-69373, Lyon, France
| | | | | | | | - Michal Guindy
- Assuta Medical Centers, Tel Aviv, Israel.,Ben Gurion University, Beersheba, Israel
| | | | | | - Xavier Castells
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | | | | | | | - Suzette Delaloge
- Institut Gustave Roussy, Villejuif, France.,Unicancer, Paris, France
| | - Sandrine de Montgolfier
- IRIS (UMR8156 CNRS & U997 INSERM), Paris 13 University, Aubervilliers, France. .,Paris Est Creteil University, Créteil, France.
| |
Collapse
|
144
|
French DP, Woof VG, Ruane H, Evans DG, Ulph F, Donnelly LS. The feasibility of implementing risk stratification into a national breast cancer screening programme: a focus group study investigating the perspectives of healthcare personnel responsible for delivery. BMC Womens Health 2022; 22:142. [PMID: 35501791 PMCID: PMC9063090 DOI: 10.1186/s12905-022-01730-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/01/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Providing women with personalized estimates of their risk of developing breast cancer, as part of routine breast cancer screening programmes, allows women at higher risk to be offered more frequent screening or drugs to reduce risk. For this to be feasible, the concept and practicalities have to be acceptable to the healthcare professionals who would put it in to practice. The present research investigated the acceptability to healthcare professionals who were responsible for the implementation of this new approach to screening in the ongoing BC-Predict study. METHODS Four focus groups were conducted with 29 healthcare professionals from a variety of professional backgrounds working within three breast screening services in north-west England. An inductive-manifest thematic analysis was conducted. RESULTS Overall, healthcare professionals viewed the implementation of personalised breast cancer risk estimation as a positive step, but discussion focused on concerns. Three major themes are presented. (1) Service constraints highlights the limited capacity within current breast services and concerns about the impact of additional workload. (2) Risk communication concerns the optimal way to convey risk to women within resource constraints. (3) Accentuating inequity discusses how risk stratification could decrease screening uptake for underserved groups. CONCLUSIONS Staff who implemented risk stratification considered it a positive addition to routine screening. They considered it essential to consider improving capacity and demands on healthcare professional time. They highlighted the need for skilled communication of risks and new pathways of care to ensure that stratification could be implemented in financially and time constrained settings without impacting negatively on women.
Collapse
Affiliation(s)
- David P French
- Division of Psychology & Mental Health, Manchester Centre for Health Psychology, School of Health Sciences, University of Manchester, Manchester, UK.
| | - Victoria G Woof
- Division of Psychology & Mental Health, Manchester Centre for Health Psychology, School of Health Sciences, University of Manchester, Manchester, UK
| | - Helen Ruane
- Nightingale & Prevent Breast Cancer Research Unit, Manchester University NHS Foundation Trust, Manchester, UK
| | - D Gareth Evans
- Nightingale & Prevent Breast Cancer Research Unit, Manchester University NHS Foundation Trust, Manchester, UK.,Division of Evolution and Genomic Science, Department of Genomic Medicine, University of Manchester, Manchester, UK
| | - Fiona Ulph
- Division of Psychology & Mental Health, Manchester Centre for Health Psychology, School of Health Sciences, University of Manchester, Manchester, UK
| | - Louise S Donnelly
- Nightingale & Prevent Breast Cancer Research Unit, Manchester University NHS Foundation Trust, Manchester, UK.,Division of Population Health, Health Services Research & Primary Care, School of Health Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
145
|
Veron L, Wehrer D, Caron O, Balleyguier C, Delaloge S. Autres approches en dépistage du cancer du sein. Bull Cancer 2022; 109:786-794. [DOI: 10.1016/j.bulcan.2022.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/24/2022] [Accepted: 02/11/2022] [Indexed: 11/26/2022]
|
146
|
Pons-Rodriguez A, Marzo-Castillejo M, Cruz-Esteve I, Galindo-Ortego G, Hernández-Leal MJ, Rué M. [Moving toward personalized breast cancer screening: The role of Primary Care]. Aten Primaria 2022; 54:102288. [PMID: 35477080 PMCID: PMC9061619 DOI: 10.1016/j.aprim.2022.102288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/11/2022] [Indexed: 11/02/2022] Open
Abstract
Breast cancer is the leading cause of death in the world among women. The Spanish National Health System (SNHS) introduced population-based breast cancer screening in 2006. As in most European programs, risk is identified on the basis of age and a mammogram is offered every two years to women aged 50-69 years. Scientific evidence is moving toward personalized screening, based on individual risk. This article presents the clinical trials that will evaluate the efficacy of personalized screening and some studies carried out in our environment on the effect of informing women of the benefits and adverse effects of screening or the acceptability and feasibility of offering personalized screening, in the Shared Decision Making context. The Preventive Activities and Health Promotion Program can help transform screening in our SNHS.
Collapse
Affiliation(s)
| | - Mercè Marzo-Castillejo
- Unitat de Suport a la Recerca Metropolitana Sud, IDIAP Jordi Gol, Direcció d'Atenció Primària Costa de Ponent, Institut Català de la Salut, Barcelona, España
| | | | | | - Maria José Hernández-Leal
- Departament d'Economia, Universitat Rovira i Virgili, Reus, España; Centre de Recerca en Economia i Sostenibilitat (ECO-SOS), Tarragona, España; Grup de Recerca en Anàlisi Estadística i Econòmica en Salut (GRAEES), Lleida y Reus, España
| | - Montserrat Rué
- Grup de Recerca en Anàlisi Estadística i Econòmica en Salut (GRAEES), Lleida y Reus, España; Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida - Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, España.
| |
Collapse
|
147
|
Noguchi N, Marinovich ML, Wylie EJ, Lund HG, Houssami N. Evidence from a BreastScreen cohort does not support a longer inter-screen interval in women who have no conventional risk factors for breast cancer. Breast 2022; 62:16-21. [PMID: 35114637 PMCID: PMC8814817 DOI: 10.1016/j.breast.2022.01.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/01/2022] Open
Abstract
Objectives To determine screening outcomes in women who have no recorded risk factors for breast cancer. Methods A retrospective population-based cohort study included all 1,026,137 mammography screening episodes in 323,082 women attending the BreastScreen Western Australia (part of national biennial screening) program between July 2007 and June 2017. Cancer detection rates (CDR) and interval cancer rates (ICR) were calculated in screening episodes with no recorded risk factors for breast cancer versus at least one risk factor stratified by age. CDR was further stratified by timeliness of screening (<27 versus ≥27 months); ICR was stratified by breast density. Results Amongst 566,948 screens (55.3%) that had no recorded risk factors, 2347 (40.9%) screen-detected cancers were observed. In screens with no risk factors, CDR was 50 (95%CI 48–52) per 10,000 screens and ICR was 7.9 (95%CI 7.4–8.4) per 10,000 women-years, estimates that were lower than screens with at least one risk factor (CDR 83 (95%CI 80–86) per 10,000 screens, ICR 12.2 (95%CI 11.5–13.0) per 10,000 women-years). Compared to timely screens with risk factors, delayed screens with no risk factors had similar CDR across all age groups and a higher proportion of node positive cancers (26.1% vs 20.7%). ICR was lowest in screens that had no risk factors nor dense breasts in all age groups. Conclusions Majority of screens had no recorded breast cancer risk factors, hence a substantial proportion of screen-detected cancers occur in these screening episodes. Our findings may not justify less frequent screening in women with no risk factors. 40.9% of screen-detected breast cancers occurred in women with no risk factors. Cancer detection rate was 50/10,000 in screens with no risk factors. Cancer size and nodal status were no more favourable in screens with no risk factors. Interval cancer rate was lowest in screens with no risk factors nor dense breasts. Our findings may not justify less frequent screening in women with no risk factors.
Collapse
Affiliation(s)
- Naomi Noguchi
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Australia.
| | | | | | | | - Nehmat Houssami
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Australia; The Daffodil Centre, The University of Sydney, Joint Venture with Cancer Council NSW, Australia
| |
Collapse
|
148
|
Peng Y, Cong Y, Lei Y, Sun F, Xu M, Zhang J, Fang L, Hong H, Cai T. Transforming Passive into Active: Multimodal Pheophytin-Based Carbon Dots Customize Protein Corona to Target Metastatic Breast Cancer. Adv Healthc Mater 2022; 11:e2102270. [PMID: 35032116 DOI: 10.1002/adhm.202102270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/28/2021] [Indexed: 12/17/2022]
Abstract
Formation of protein corona on nanomaterials surface in vivo is usually considered as an unpredictable event for a predefined targeted delivery system for malignant cancers. In most situations, these protein coronas substantially change targeting efficiency or even cause adverse reactions which both hinder the clinical translation of the cargo-delivery systems. Active customization of protein corona onto nanomaterials surfaces can benefit their biomedical performances and open up new opportunities in construction of targeted delivery systems. Herein, lipid-PEG/pheophytin carbon dots (LPCDs) are prepared from natural chlorophyll and integrate seamlessly with positron emission tomography imaging, near-infrared fluorescence imaging, and photodynamic therapy capacity. In vitro measurements demonstrate that the LPCDs can actively absorb apolipoproteins into the protein corona to enhance their uptakes in breast cancer cells. In vivo studies confirm that LPCDs can give accurate delineation of metastatic breast cancer foci from surrounding normal tissues with multimodal biomedical functions. The feasibility of using LPCDs as a multimodal imaging and cancer-targeting nanoplatform may provide impetus for developing precise yet facile protein corona-targeted delivery systems for future clinical practice.
Collapse
Affiliation(s)
- Yayun Peng
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 China
| | - Yiyang Cong
- State Key Laboratory of Pharmaceutical Biotechnology Jiangsu Key Laboratory of Molecular Medicine School of Medicine Medical School of Nanjing University Nanjing 210093 China
| | - Yuzhu Lei
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 China
| | - Fanwen Sun
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 China
| | - Menghan Xu
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 China
| | - Jingzi Zhang
- Jiangsu Key Laboratory of Molecular Medicine Chemistry and Biomedicine Innovation Center Medical School of Nanjing University Nanjing 210093 China
| | - Lei Fang
- Jiangsu Key Laboratory of Molecular Medicine Chemistry and Biomedicine Innovation Center Medical School of Nanjing University Nanjing 210093 China
| | - Hao Hong
- State Key Laboratory of Pharmaceutical Biotechnology Jiangsu Key Laboratory of Molecular Medicine School of Medicine Medical School of Nanjing University Nanjing 210093 China
| | - Ting Cai
- State Key Laboratory of Natural Medicines Department of Pharmaceutics China Pharmaceutical University Nanjing 210009 China
| |
Collapse
|
149
|
Fitzgerald RC, Antoniou AC, Fruk L, Rosenfeld N. The future of early cancer detection. Nat Med 2022; 28:666-677. [PMID: 35440720 DOI: 10.1038/s41591-022-01746-x] [Citation(s) in RCA: 145] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/15/2022] [Indexed: 12/22/2022]
Abstract
A proactive approach to detecting cancer at an early stage can make treatments more effective, with fewer side effects and improved long-term survival. However, as detection methods become increasingly sensitive, it can be difficult to distinguish inconsequential changes from lesions that will lead to life-threatening cancer. Progress relies on a detailed understanding of individualized risk, clear delineation of cancer development stages, a range of testing methods with optimal performance characteristics, and robust evaluation of the implications for individuals and society. In the future, advances in sensors, contrast agents, molecular methods, and artificial intelligence will help detect cancer-specific signals in real time. To reduce the burden of cancer on society, risk-based detection and prevention needs to be cost effective and widely accessible.
Collapse
Affiliation(s)
- Rebecca C Fitzgerald
- Early Detection Programme, Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, UK.
| | - Antonis C Antoniou
- Centre for Cancer Genetic Epidemiology, Department of Public Health & Primary Care, University of Cambridge, Cambridge, UK
| | - Ljiljana Fruk
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| |
Collapse
|
150
|
Clift AK, Hippisley-Cox J, Dodwell D, Lord S, Brady M, Petrou S, Collins GS. Development and validation of clinical prediction models for breast cancer incidence and mortality: a protocol for a dual cohort study. BMJ Open 2022; 12:e050828. [PMID: 35351695 PMCID: PMC8961149 DOI: 10.1136/bmjopen-2021-050828] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 01/07/2022] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Breast cancer is the most common cancer and the leading cause of cancer-related death in women worldwide. Risk prediction models may be useful to guide risk-reducing interventions (such as pharmacological agents) in women at increased risk or inform screening strategies for early detection methods such as screening. METHODS AND ANALYSIS The study will use data for women aged 20-90 years between 2000 and 2020 from QResearch linked at the individual level to hospital episodes, cancer registry and death registry data. It will evaluate a set of modelling approaches to predict the risk of developing breast cancer within the next 10 years, the 'combined' risk of developing a breast cancer and then dying from it within 10 years, and the risk of breast cancer mortality within 10 years of diagnosis. Cox proportional hazards, competing risks, random survival forest, deep learning and XGBoost models will be explored. Models will be developed on the entire dataset, with 'apparent' performance reported, and internal-external cross-validation used to assess performance and geographical and temporal transportability (two 10-year time periods). Random effects meta-analysis will pool discrimination and calibration metric estimates from individual geographical units obtained from internal-external cross-validation. We will then externally validate the models in an independent dataset. Evaluation of performance heterogeneity will be conducted throughout, such as exploring performance across ethnic groups. ETHICS AND DISSEMINATION Ethics approval was granted by the QResearch scientific committee (reference number REC 18/EM/0400: OX129). The results will be written up for submission to peer-reviewed journals.
Collapse
Affiliation(s)
- Ashley Kieran Clift
- Cancer Research UK Oxford Centre, University of Oxford, Oxford, UK
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Julia Hippisley-Cox
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - David Dodwell
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Simon Lord
- Department of Oncology, University of Oxford, Oxford, UK
| | - Mike Brady
- Department of Oncology, University of Oxford, Oxford, UK
| | - Stavros Petrou
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Gary S Collins
- Centre for Statistics in Medicine, University of Oxford, Oxford, UK
| |
Collapse
|