1
|
Austin D, Maier RH, Akhter N, Sayari M, Ogundimu E, Maddox JM, Vahabi S, Humphreys AC, Graham J, Oxenham H, Haney S, Cresti N, Verrill M, Osborne W, Wright KL, Goranova R, Bailey JR, Kalakonda N, Macheta M, Kilner MF, Young ME, Morley NJ, Neelakantan P, Gilbert G, Thomas BK, Graham RJ, Fujisawa T, Mills NL, Hildreth V, Prichard J, Kasim AS, Hancock HC, Plummer C. Preventing Cardiac Damage in Patients Treated for Breast Cancer and Lymphoma: The PROACT Clinical Trial. JACC CardioOncol 2024; 6:684-696. [PMID: 39479329 PMCID: PMC11520224 DOI: 10.1016/j.jaccao.2024.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 11/02/2024] Open
Abstract
Background Cardiotoxicity is a concern for cancer survivors undergoing anthracycline chemotherapy. Enalapril has been explored for its potential to mitigate cardiotoxicity in cancer patients. The dose-dependent cardiotoxicity effects of anthracyclines can be detected early through the biomarker cardiac troponin. Objectives The PROACT (Preventing Cardiac Damage in Patients Treated for Breast Cancer and Lymphoma) clinical trial assessed the effectiveness of enalapril in preventing cardiotoxicity, manifesting as myocardial injury and cardiac function impairment, in patients undergoing high-dose anthracycline-based chemotherapy for breast cancer or non-Hodgkin lymphoma. Methods This prospective, multicenter, open-label, randomized controlled trial employed a superiority design with observer-blinded endpoints. A total of 111 participants, scheduled for 6 cycles of chemotherapy with a planned dose of ≥300 mg/m2 doxorubicin equivalents, were randomized to receive either enalapril (titrated up to 20 mg daily) or standard care without enalapril. Results Myocardial injury, indicated by cardiac troponin T (≥14 ng/L), during and 1 month after chemotherapy, was observed in 42 (77.8%) of 54 patients in the enalapril group vs 45 (83.3%) of 54 patients in the standard care group (OR: 0.65; 95% CI: 0.23-1.78). Injury detected by cardiac troponin I (>26.2 ng/L) occurred in 25 (47.2%) of 53 patients on enalapril compared with 24 (45.3%) of 53 in standard care (OR: 1.10; 95% CI: 0.50-2.38). A relative decline of more than 15% from baseline in left ventricular global longitudinal strain was observed in 10 (21.3%) of 47 patients on enalapril and 9 (21.9%) of 41 in standard care (OR: 0.95; 95% CI: 0.33-2.74). An absolute decline of >10% to <50% in left ventricular ejection fraction was seen in 2 (4.1%) of 49 patients on enalapril vs none in patients in standard care. Conclusions Adding enalapril to standard care during chemotherapy did not prevent cardiotoxicity in patients receiving high-dose anthracycline-based chemotherapy. (PROACT: Can we prevent Chemotherapy-related Heart Damage in Patients With Breast Cancer and Lymphoma?; NCT03265574).
Collapse
Affiliation(s)
- David Austin
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rebecca H. Maier
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nasima Akhter
- School of Health and Life Sciences, Teesside University, Middlesbrough, United Kingdom
| | - Mohammad Sayari
- Department of Mathematical Science, Durham University, Durham, United Kingdom
| | - Emmanuel Ogundimu
- Department of Mathematical Science, Durham University, Durham, United Kingdom
| | - Jamie M. Maddox
- Department of Haematology, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Sharareh Vahabi
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Alison C. Humphreys
- Cancer Services, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Janine Graham
- Cancer Services, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Cancer Services, North Tees and Hartlepool NHS Foundation Trust, Stockton-on-Tees, United Kingdom
| | - Helen Oxenham
- Department of Cardiology, North Tees and Hartlepool NHS Foundation Trust, Stockton-on-Tees, United Kingdom
| | - Sophie Haney
- Cancer Services, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Cancer Services, County Durham and Darlington NHS Foundation Trust, Darlington, United Kingdom
| | - Nicola Cresti
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Mark Verrill
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Wendy Osborne
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Kathryn L. Wright
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
- Cancer Services, South Tyneside and Sunderland NHS Foundation Trust, Sunderland, United Kingdom
| | - Rebecca Goranova
- Department of Oncology, Derriford Hospital, University Hospitals Plymouth NHS Trust, Plymouth, United Kingdom
| | - James R. Bailey
- Department of Haematology, Hull University Teaching Hospitals NHS Trust, Queen’s Centre for Oncology and Haematology, Castle Hill Hospital, Hull, United Kingdom
| | - Nagesh Kalakonda
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Mac Macheta
- Department of Haematology, Blackpool Teaching Hospitals, Blackpool, United Kingdom
| | - Mari F. Kilner
- Pathology Services, Northumbria Healthcare NHS Foundation Trust, North Shields, United Kingdom
| | - Moya E. Young
- Department of Haematology, East Kent Hospitals University NHS Foundation Trust, Canterbury, United Kingdom
| | - Nick J. Morley
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Pratap Neelakantan
- Department of Haematology, Royal Berkshire NHS Foundation Trust, Reading, United Kingdom
| | - Georgia Gilbert
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Byju K. Thomas
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Richard J. Graham
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Takeshi Fujisawa
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicholas L. Mills
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Victoria Hildreth
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jonathan Prichard
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Adetayo S. Kasim
- Department of Anthropology, Durham University, Durham, United Kingdom
| | - Helen C. Hancock
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Chris Plummer
- Department of Cardiology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
2
|
Ohnstad HO, Blix ES, Akslen LA, Gilje B, Raj SX, Skjerven H, Borgen E, Janssen EAM, Mortensen E, Brekke MB, Falk RS, Schlichting E, Boge B, Songe-Møller S, Olsson P, Heie A, Mannsåker B, Vestlid MA, Kursetgjerde T, Gravdehaug B, Suhrke P, Sanchez E, Bublevic J, Røe OD, Geitvik GA, Halset EH, Rypdal MC, Langerød A, Lømo J, Garred Ø, Porojnicu A, Engebraaten O, Geisler J, Lyngra M, Hansen MH, Søiland H, Nakken T, Asphaug L, Kristensen V, Sørlie T, Nygård JF, Kiserud CE, Reinertsen KV, Russnes HG, Naume B. Impact of Prosigna test on adjuvant treatment decision in lymph node-negative early breast cancer-a prospective national multicentre study (EMIT-1). ESMO Open 2024; 9:103475. [PMID: 38838499 PMCID: PMC11190479 DOI: 10.1016/j.esmoop.2024.103475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND EMIT-1 is a national, observational, single-arm trial designed to assess the value of the Prosigna, Prediction Analysis of Microarray using the 50 gene classifier (PAM50)/Risk of Recurrence (ROR), test as a routine diagnostic tool, examining its impact on adjuvant treatment decisions, clinical outcomes, side-effects and cost-effectiveness. Here we present the impact on treatment decisions. PATIENTS AND METHODS Patients with hormone receptor-positive, human epidermal growth factor receptor 2-negative pT1-pT2 lymph node-negative early breast cancer (EBC) were included. The Prosigna test and standard histopathology assessments were carried out. Clinicians' treatment decisions were recorded before (pre-Prosigna) and after (post-Prosigna) the Prosigna test results were disclosed. RESULTS Of 2217 patients included, 2178 had conclusive Prosigna results. The pre-Prosigna treatment decisions were: no systemic treatment (NT) in 27% of patients, endocrine treatment alone (ET) in 38% and chemotherapy (CT) followed by ET (CT + ET) in 35%. Post-Prosigna treatment decisions were 25% NT, 51% ET and 24% CT + ET, respectively. Adjuvant treatment changed in 28% of patients, including 21% change in CT use. Among patients assigned to CT + ET pre-Prosigna, 45% were de-escalated to ET post-Prosigna. Of patients assigned to ET, 12% were escalated to CT + ET and 8% were de-escalated to NT; of those assigned to NT, 18% were escalated to ET/CT + ET. CT was more frequently recommended for patients aged ≤50 years. In the subgroup with pT1c-pT2 G2 and intermediate Ki67 (0.5-1.5× local laboratory median Ki67 score), the pre-Prosigna CT treatment decision varied widely across hospitals (3%-51%). Post-Prosigna, the variability of CT use was markedly reduced (8%-24%). The correlation between Ki67 and ROR score within this subgroup was poor (r = 0.25-0.39). The median ROR score increased by increasing histological grade, but the ROR score ranges were wide (for G1 0-79, G2 0-90, G3 16-94). CONCLUSION The Prosigna test result changed adjuvant treatment decisions in all EBC clinical risk groups, markedly decreased the CT use for patients categorized as higher clinical risk pre-Prosigna and reduced treatment decision discrepancies between hospitals.
Collapse
Affiliation(s)
- H O Ohnstad
- Department of Oncology, Division of Cancer Medicine, Oslo University Hospital, Oslo
| | - E S Blix
- Department of Oncology, University of North Norway, Tromsø; Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø
| | - L A Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, Section for Pathology, University of Bergen, Bergen; Department of Pathology Haukeland University Hospital, Bergen
| | - B Gilje
- Department of Haematology and Oncology, Stavanger University Hospital, Stavanger
| | - S X Raj
- Department of Oncology, St Olavs Hospital, Trondheim
| | - H Skjerven
- Department of Breast Surgery, Vestre Viken Hospital Trust, Drammen
| | - E Borgen
- Department of Pathology, Division of Laboratory Medicine, Oslo University Hospital, Oslo
| | - E A M Janssen
- Department of Pathology, Stavanger University Hospital, Stavanger; Department of Chemistry, Bioscience and Environmental Engineering, Stavanger University, Stavanger, Norway; Menzies Health Institute Queensland and Griffith University, Southport, Australia
| | - E Mortensen
- Department of Pathology, University of North Norway, Tromsø
| | - M B Brekke
- Department of Pathology, St Olavs Hospital, Trondheim
| | - R S Falk
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo
| | - E Schlichting
- Department of Oncology, Breast and Endocrine Surgery Unit, Division of Cancer Medicine, Oslo University Hospital, Oslo
| | - B Boge
- Department of Oncology, Hospital of Southern Norway, Kristiansand
| | | | - P Olsson
- Department of Breast Surgery, Innlandet Hospital Trust, Hamar
| | - A Heie
- Department of Breast Surgery, Haukeland University Hospital, Bergen
| | - B Mannsåker
- Department of Oncology, Nordland Hospital, Bodø
| | - M A Vestlid
- Department of Breast Surgery, Telemark Hospital Trust, Skien
| | - T Kursetgjerde
- Department of Oncology, Møre og Romsdal Hospital Trust, Ålesund
| | - B Gravdehaug
- Department of Breast Surgery, Akershus University Hospital, Lørenskog
| | - P Suhrke
- Department of Pathology, Vestfold Hospital Trust, Tønsberg
| | - E Sanchez
- Department of Oncology, Haugesund Hospital, Haugesund
| | - J Bublevic
- Department of Oncology, Førde Central Hospital, Førde
| | - O D Røe
- Department of Oncology, Levanger Hospital, Levanger
| | - G A Geitvik
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo
| | - E H Halset
- Department of Oncology, Division of Cancer Medicine, Oslo University Hospital, Oslo
| | - M C Rypdal
- Department of Pathology, Division of Laboratory Medicine, Oslo University Hospital, Oslo
| | - A Langerød
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo
| | - J Lømo
- Department of Pathology, Division of Laboratory Medicine, Oslo University Hospital, Oslo
| | - Ø Garred
- Department of Pathology, Division of Laboratory Medicine, Oslo University Hospital, Oslo
| | - A Porojnicu
- Department of Oncology, Vestre Viken Hospital Trust, Drammen
| | - O Engebraaten
- Department of Oncology, Division of Cancer Medicine, Oslo University Hospital, Oslo; Institute of Clinical Medicine, University of Oslo, Oslo
| | - J Geisler
- Institute of Clinical Medicine, University of Oslo, Oslo; Department of Oncology, Akershus University Hospital, Lørenskog
| | - M Lyngra
- Department of Pathology, Akershus University Hospital, Lørenskog
| | - M H Hansen
- Department of Breast Surgery, University of North Norway, Tromsø
| | - H Søiland
- Department of Research, Stavanger University Hospital, Stavanger; Department of Clinical Science, University of Bergen, Bergen
| | - T Nakken
- User representative, Oslo University Hospital, Oslo
| | - L Asphaug
- Clinical Trials Unit, Oslo University Hospital, Oslo; Institute of Health and Society, Faculty of Medicine, University of Oslo, Oslo
| | - V Kristensen
- Institute of Clinical Medicine, University of Oslo, Oslo
| | - T Sørlie
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo; Institute of Clinical Medicine, University of Oslo, Oslo
| | | | - C E Kiserud
- National Advisory Unit for Late Effects after Cancer Treatment, Oslo University Hospital, Oslo, Norway
| | - K V Reinertsen
- Department of Oncology, Division of Cancer Medicine, Oslo University Hospital, Oslo; National Advisory Unit for Late Effects after Cancer Treatment, Oslo University Hospital, Oslo, Norway
| | - H G Russnes
- Department of Pathology, Division of Laboratory Medicine, Oslo University Hospital, Oslo; Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo; Institute of Clinical Medicine, University of Oslo, Oslo
| | - B Naume
- Department of Oncology, Division of Cancer Medicine, Oslo University Hospital, Oslo; Institute of Clinical Medicine, University of Oslo, Oslo.
| |
Collapse
|
3
|
Peters AL, Hall PS, Jordan LB, Soh FY, Hannington L, Makaranka S, Urquhart G, Vallet M, Cartwright D, Marashi H, Elsberger B. Enhancing clinical decision support with genomic tools in breast cancer: A Scottish perspective. Breast 2024; 75:103728. [PMID: 38657322 PMCID: PMC11061332 DOI: 10.1016/j.breast.2024.103728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 03/12/2024] [Accepted: 04/03/2024] [Indexed: 04/26/2024] Open
Abstract
INTRODUCTION The Oncotype DX Breast RS test has been adopted in Scotland and has been the subject of a large population-based study by a Scottish Consensus Group to assess the uptake of the recurrence score (RS), evaluate co-variates associated with the RS and to analyse the effect it may have had on clinical practice. MATERIALS & METHODS Pan-Scotland study between August 2018-August 2021 evaluating 833 patients who had a RS test performed as part of their diagnostic pathway. Data was extracted retrospectively from electronic records and analysis conducted to describe change in chemotherapy administration (by direct comparison with conventional risk assessment tools), and univariate/multivariate analysis to assess relationship between covariates and the RS. RESULTS Chemotherapy treatment was strongly influenced by the RS (p < 0.001). Only 30 % of patients received chemotherapy treatment in the intermediate and high risk PREDICT groups, where chemotherapy is considered. Additionally, 55.5 % of patients with a high risk PREDICT had a low RS and did not receive chemotherapy. There were 17 % of patients with a low risk PREDICT but high RS who received chemotherapy. Multivariate regression analysis showed the progesterone receptor Allred score (PR score) to be a strong independent predictor of the RS, with a negative PR score being associated with high RS (OR 4.49, p < 0.001). Increasing grade was also associated with high RS (OR 3.81, p < 0.001). Classic lobular pathology was associated with a low RS in comparison to other tumour pathology (p < 0.01). Nodal disease was associated with a lower RS (p = 0.012) on univariate analysis, with menopausal status (p = 0.43) not influencing the RS on univariate or multivariate analysis. CONCLUSIONS Genomic assays offer the potential for risk-stratified decision making regarding the use of chemotherapy. They can help reduce unnecessary chemotherapy treatment and identify a subgroup of patients with more adverse genomic tumour biology. A recent publication by Health Improvement Scotland (HIS) has updated guidance on use of the RS test for NHS Scotland. It suggests to limit its use to the intermediate risk PREDICT group. Our study shows the impact of the RS test in the low and high risk PREDICT groups. The implementation across Scotland has resulted in a notable shift in practice, leading to a significant reduction in chemotherapy administration in the setting of high risk PREDICT scores returning low risk RS. There has also been utility for the test in the low risk PREDICT group to detect a small subgroup with a high RS. We have found the PR score to have a strong independent association with high risk RS. This finding was not evaluated by the key RS test papers, and the potential prognostic information provided by the PR score as a surrogate biomarker is an outstanding question that requires more research to validate.
Collapse
Affiliation(s)
- A L Peters
- Beatson West of Scotland Cancer Centre, Gartnavel Hospital, NHS Greater Glasgow & Clyde, 1053 Great Western Rd, Glasgow G12 0YN, UK; Cancer Research UK (CRUK) Scotland Institute, Switchback Road, Bearsden, Glasgow G61 1BD, UK.
| | - P S Hall
- Edinburgh Cancer Research Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - L B Jordan
- Ninewells Hospital & Medical School, NHS Tayside, Department of Pathology, Dundee, DD1 9SY, UK
| | - F Y Soh
- Raigmore Hospital, NHS Highland, Department of Oncology, Inverness IV2 3UJ, UK
| | - L Hannington
- Beatson West of Scotland Cancer Centre, Gartnavel Hospital, NHS Greater Glasgow & Clyde, 1053 Great Western Rd, Glasgow G12 0YN, UK
| | - S Makaranka
- Aberdeen Royal Infirmary, NHS Grampian, Department of Breast Surgery, Aberdeen AB25 2ZN, UK
| | - G Urquhart
- Aberdeen Royal Infirmary, NHS Grampian, Department of Oncology, Aberdeen AB25 2ZN, UK
| | - M Vallet
- Edinburgh Cancer Research Centre, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - D Cartwright
- Beatson West of Scotland Cancer Centre, Gartnavel Hospital, NHS Greater Glasgow & Clyde, 1053 Great Western Rd, Glasgow G12 0YN, UK; Cancer Research UK (CRUK) Scotland Institute, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - H Marashi
- Beatson West of Scotland Cancer Centre, Gartnavel Hospital, NHS Greater Glasgow & Clyde, 1053 Great Western Rd, Glasgow G12 0YN, UK
| | - B Elsberger
- Aberdeen Royal Infirmary, NHS Grampian, Department of Breast Surgery, Aberdeen AB25 2ZN, UK
| |
Collapse
|
4
|
Venetis K, Pescia C, Cursano G, Frascarelli C, Mane E, De Camilli E, Munzone E, Dellapasqua S, Criscitiello C, Curigliano G, Guerini Rocco E, Fusco N. The Evolving Role of Genomic Testing in Early Breast Cancer: Implications for Diagnosis, Prognosis, and Therapy. Int J Mol Sci 2024; 25:5717. [PMID: 38891906 PMCID: PMC11172282 DOI: 10.3390/ijms25115717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Multigene prognostic genomic assays have become indispensable in managing early breast cancer (EBC), offering crucial information for risk stratification and guiding adjuvant treatment strategies in conjunction with traditional clinicopathological parameters. The American Society of Clinical Oncology (ASCO) guidelines endorse these assays, though some clinical contexts still lack definitive recommendations. The dynamic landscape of EBC management demands further refinement and optimization of genomic assays to streamline their incorporation into clinical practice. The breast cancer community is poised at the brink of transformative advances in enhancing the clinical utility of genomic assays, aiming to significantly improve the precision and effectiveness of both diagnosis and treatment for women with EBC. This article methodically examines the testing methodologies, clinical validity and utility, costs, diagnostic frameworks, and methodologies of the established genomic tests, including the Oncotype Dx Breast Recurrence Score®, MammaPrint, Prosigna®, EndoPredict®, and Breast Cancer Index (BCI). Among these tests, Prosigna and EndoPredict® have at present been validated only on a prognostic level, while Oncotype Dx, MammaPrint, and BCI hold both a prognostic and predictive role. Oncologists and pathologists engaged in the management of EBC will find in this review a thorough comparison of available genomic assays, as well as strategies to optimize the utilization of the information derived from them.
Collapse
Affiliation(s)
- Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
| | - Carlo Pescia
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- School of Pathology, University of Milan, 20122 Milan, Italy
| | - Giulia Cursano
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| | - Chiara Frascarelli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| | - Eltjona Mane
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
| | - Elisa De Camilli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
| | - Elisabetta Munzone
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.M.); (S.D.)
| | - Silvia Dellapasqua
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.M.); (S.D.)
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
- Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
- Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Elena Guerini Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| |
Collapse
|
5
|
Rojas L, Rojas-Reyes MX, Rosselli D, Ariza JG, Ruiz-Patiño A, Cardona AF. Cost-utility analysis of genomic profiling in early breast cancer in Colombia. COST EFFECTIVENESS AND RESOURCE ALLOCATION 2023; 21:42. [PMID: 37430303 DOI: 10.1186/s12962-023-00449-5] [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: 11/08/2022] [Accepted: 06/14/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND In Colombia, the best strategy to establish indication for adjuvant chemotherapy in early breast cancer (EBC) remains unknown. This study aimed to identify the cost-utility of Oncotype DX™ (ODX) or Mammaprint™ (MMP) tests to establish the necessity of adjuvant chemotherapy. METHODS This study used an adapted decision-analytic model to compare cost and outcomes of care between ODX or MMP tests and routine care without ODX or MMP tests (adjuvant chemotherapy for all patients) over a 5-year time horizon from the perspective of the Colombian National Health System (NHS; payer). Inputs were obtained from national unit cost tariffs, published literature, and clinical trial database. The study population comprised women with hormone-receptor-positive (HR +), HER2-negative, lymph-node-negative (LN0) EBC with high-risk clinical criteria for recurrence. The outcome measures were discounted incremental cost-utility ratio (ICUR; 2021 United States dollar per quality-adjusted life-year [QALY] gained) and net monetary benefit (NMB). Probabilistic (PSA) and deterministic sensitivity analysis (DSA) were performed. RESULTS ODX increases QALYs by 0.05 and MMP by 0.03 with savings of $2374 and $554 compared with the standard strategy, respectively, and were cost-saving in cost-utility plane. NMB for ODX was $2203 and for MMP was $416. Both tests dominate the standard strategy. Sensitivity analysis revealed that with a threshold of 1 gross domestic product per capita, ODX will be cost-effective in 95.5% of the cases compared with 70.2% cases involving MMP.DSA showed that the variable with significant influence was the monthly cost of adjuvant chemotherapy. PSA revealed that ODX was a consistently superior strategy. CONCLUSIONS Genomic profiling using ODX or MMP tests to define the need of adjuvant chemotherapy treatment in patients with HR + and HER2 -EBC is a cost-effective strategy that allows Colombian NHS to maintain budget.
Collapse
Affiliation(s)
- Leonardo Rojas
- Thoracic and GU Unit, Fundación Centro de Tratamiento en Investigación Sobre Cáncer Luis Carlos Sarmiento Angulo (CTIC), Carrera 14 # 169 -49, Office 204, Bogotá, Colombia.
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia.
| | | | - Diego Rosselli
- Department of Clinical Epidemiology and Biostatistics, Faculty of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | | | - Andrés F Cardona
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
- Foundation for Clinical and Applied Cancer Research-FICMAC, Bogotá, Colombia
- Direction of Research, Science and Education, Fundación Centro de Tratamiento en Investigación Sobre Cáncer Luis Carlos Sarmiento Angulo (CTIC), Bogotá, Colombia
| |
Collapse
|
6
|
Neves Rebello Alves L, Dummer Meira D, Poppe Merigueti L, Correia Casotti M, do Prado Ventorim D, Ferreira Figueiredo Almeida J, Pereira de Sousa V, Cindra Sant'Ana M, Gonçalves Coutinho da Cruz R, Santos Louro L, Mendonça Santana G, Erik Santos Louro T, Evangelista Salazar R, Ribeiro Campos da Silva D, Stefani Siqueira Zetum A, Silva Dos Reis Trabach R, Imbroisi Valle Errera F, de Paula F, de Vargas Wolfgramm Dos Santos E, Fagundes de Carvalho E, Drumond Louro I. Biomarkers in Breast Cancer: An Old Story with a New End. Genes (Basel) 2023; 14:1364. [PMID: 37510269 PMCID: PMC10378988 DOI: 10.3390/genes14071364] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Breast cancer is the second most frequent cancer in the world. It is a heterogeneous disease and the leading cause of cancer mortality in women. Advances in molecular technologies allowed for the identification of new and more specifics biomarkers for breast cancer diagnosis, prognosis, and risk prediction, enabling personalized treatments, improving therapy, and preventing overtreatment, undertreatment, and incorrect treatment. Several breast cancer biomarkers have been identified and, along with traditional biomarkers, they can assist physicians throughout treatment plan and increase therapy success. Despite the need of more data to improve specificity and determine the real clinical utility of some biomarkers, others are already established and can be used as a guide to make treatment decisions. In this review, we summarize the available traditional, novel, and potential biomarkers while also including gene expression profiles, breast cancer single-cell and polyploid giant cancer cells. We hope to help physicians understand tumor specific characteristics and support decision-making in patient-personalized clinical management, consequently improving treatment outcome.
Collapse
Affiliation(s)
- Lyvia Neves Rebello Alves
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória 29047-105, ES, Brazil
| | - Débora Dummer Meira
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória 29047-105, ES, Brazil
| | - Luiza Poppe Merigueti
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
| | - Matheus Correia Casotti
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória 29047-105, ES, Brazil
| | - Diego do Prado Ventorim
- Instituto Federal de Educação, Ciência e Tecnologia do Espírito Santo (Ifes), Cariacica 29150-410, ES, Brazil
| | - Jucimara Ferreira Figueiredo Almeida
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
| | - Valdemir Pereira de Sousa
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória 29047-105, ES, Brazil
| | - Marllon Cindra Sant'Ana
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
| | - Rahna Gonçalves Coutinho da Cruz
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
| | - Luana Santos Louro
- Centro de Ciências da Saúde, Curso de Medicina, Universidade Federal do Espírito Santo (UFES), Vitória 29090-040, ES, Brazil
| | - Gabriel Mendonça Santana
- Centro de Ciências da Saúde, Curso de Medicina, Universidade Federal do Espírito Santo (UFES), Vitória 29090-040, ES, Brazil
| | - Thomas Erik Santos Louro
- Escola Superior de Ciências da Santa Casa de Misericórdia de Vitória (EMESCAM), Vitória 29027-502, ES, Brazil
| | - Rhana Evangelista Salazar
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória 29047-105, ES, Brazil
| | - Danielle Ribeiro Campos da Silva
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória 29047-105, ES, Brazil
| | - Aléxia Stefani Siqueira Zetum
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória 29047-105, ES, Brazil
| | - Raquel Silva Dos Reis Trabach
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
| | - Flávia Imbroisi Valle Errera
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória 29047-105, ES, Brazil
| | - Flávia de Paula
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória 29047-105, ES, Brazil
| | - Eldamária de Vargas Wolfgramm Dos Santos
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória 29047-105, ES, Brazil
| | - Elizeu Fagundes de Carvalho
- Instituto de Biologia Roberto Alcântara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro 20551-030, RJ, Brazil
| | - Iúri Drumond Louro
- Núcleo de Genética Humana e Molecular, Departamento de Ciências Biológicas, Universidade Federal do Espírito Santo (UFES), Vitória 29075-910, ES, Brazil
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória 29047-105, ES, Brazil
| |
Collapse
|
7
|
Xu H, Li L, Qu L, Tu J, Sun X, Liu X, Xu K. Atractylenolide-1 affects glycolysis/gluconeogenesis by downregulating the expression of TPI1 and GPI to inhibit the proliferation and invasion of human triple-negative breast cancer cells. Phytother Res 2023; 37:820-833. [PMID: 36420870 DOI: 10.1002/ptr.7661] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/25/2022]
Abstract
Atractylenolide-1 (AT-1) is a major octanol alkaloid isolated from Atractylodes Rhizoma and is widely used to treat various diseases. However, few reports have addressed the anticancer potential of AT-1, and the underlying molecular mechanisms of its anticancer effects are unclear. This study aimed to assess the effect of AT-1 on triple-negative breast cancer (TNBC) cell proliferation and migration and explore its potential molecular mechanisms. Cell invasion assays confirmed that the number of migrating cells decreased after AT-1 treatment. Colony formation assays showed that AT-1 treatment impaired the ability of MDA-MB-231 cells to form colonies. AT-1 inhibited the expression of p-p38, p-ERK, and p-AKT in MDA-MB-231 cells, significantly downregulated the proliferation of anti-apoptosis-related proteins CDK1, CCND1, and Bcl2, and up-regulated pro-apoptotic proteins Bak, caspase 3, and caspase 9. The gas chromatography-mass spectroscopy results showed that AT-1 downregulated the metabolism-related genes TPI1 and GPI through the glycolysis/gluconeogenesis pathway and inhibited tumor growth in vivo. AT-1 affected glycolysis/gluconeogenesis by downregulating the expression of TPI1 and GPI, inhibiting the proliferation, migration, and invasion of (TNBC) MDA-MB-231 cells and suppressing tumor growth in vivo.
Collapse
Affiliation(s)
- Haiying Xu
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Lanqing Li
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Linghang Qu
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Jiyuan Tu
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Xiongjie Sun
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Xianqiong Liu
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Kang Xu
- Hubei Engineering Technology Research Center of Chinese Materia Medica Processing, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
8
|
O'Shaughnessy J, Gradishar W, O'Regan R, Gadi V. Risk of Recurrence in Patients with HER2+ Early-Stage Breast Cancer: Literature Analysis of Patient and Disease Characteristics. Clin Breast Cancer 2023; 23:350-362. [PMID: 37149421 DOI: 10.1016/j.clbc.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/10/2023] [Accepted: 03/18/2023] [Indexed: 03/31/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) overexpression occurs in 15% to 20% of patients with early-stage breast cancers (EBCs). Without HER2-targeted therapy, 30% to 50% of patients relapse within 10 years, many developing incurable metastatic disease. This literature review was designed to identify and validate patient- and disease-related factors associated with recurrence in patients with HER2+ EBC. Peer-reviewed primary research articles and congress abstracts were identified by searching MEDLINE. Articles published in English from 2019 to 2022 were included to identify contemporary treatment options. Results were analyzed for the relationship between risk factors and surrogates of HER2+ EBC recurrence to determine how identified risk factors affected HER2+ EBC recurrence. Sixty-one articles and 65 abstracts that assessed age at diagnosis, body mass index (BMI), tumor size at diagnosis, hormone receptor (HR) status, pathologic complete response (pCR) status, and biomarkers were analyzed. We confirmed the results of previously published reviews reporting residual cancer burden >0, non-pCR, and fewer tumor-infiltrating lymphocytes (TILs) as risk factors of recurrence. HR status remained an important risk factor for recurrence, with HER2+/HR+ disease more likely to recur. Two or more positive lymph nodes, higher BMI, larger primary tumor size, and low Ki67 were more commonly associated with HER2+ EBC recurrence. The identification of patient and disease factors frequently associated with HER2+ EBC recurrence in the literature provides insight into potential recurrence risk factors. Further investigation into the risk factors identified in this review could lead to improved treatments for patients at high risk for HER2+ EBC recurrence.
Collapse
Affiliation(s)
- Joyce O'Shaughnessy
- Baylor University Medical Center, Dallas, TX; Texas Oncology-Baylor Charles A. Sammons Cancer Center, Dallas, TX.
| | | | - Ruth O'Regan
- Department of Medicine, University of Rochester, Rochester, NY
| | - Vijayakrishna Gadi
- Department of Medicine, University of Illinois Chicago, Chicago, IL; Translational Oncology Program, University of Illinois Cancer Center, Chicago, IL
| |
Collapse
|
9
|
Curigliano G, Cardoso F, Gnant M, Harbeck N, King J, Laenkholm AV, Penault-Llorca F, Prat A. PROCURE European consensus on breast cancer multigene signatures in early breast cancer management. NPJ Breast Cancer 2023; 9:8. [PMID: 36828834 PMCID: PMC9951144 DOI: 10.1038/s41523-023-00510-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 01/26/2023] [Indexed: 02/26/2023] Open
Abstract
Breast cancer multigene signatures (BCMS) have changed how patients with early-stage breast cancer (eBC) are managed, as they provide prognostic information and can be used to select patients who may avoid adjuvant chemotherapy. Clinical guidelines make recommendations on the use of BCMS; however, little is known on the current use of BCMS in clinical practice. We conduct a two-round Delphi survey to enquire about current use and perceived utility for specific patient profiles, and unmet needs of BCMS. Overall, 133 panellists experienced in breast cancer across 11 European countries have participated, most using BCMS either routinely (66.2%) or in selected cases (27.1%). Our results show that BCMS are mainly used to assess the risk of recurrence and to select patients for adjuvant chemotherapy; notably, no consensus has been reached on the lack of utility of BCMS for selecting the type of chemotherapy to administer. Also, there are discrepancies between the recommended and current use of BCMS in clinical practice, with use in certain patient profiles for which there is no supporting evidence. Our study suggests that physician education initiatives are needed to ensure the correct use and interpretation of BCMS to, ultimately, improve management of patients with eBC.
Collapse
Affiliation(s)
- Giuseppe Curigliano
- European Institute of Oncology, IRCCS, Milan, Italy. .,Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy.
| | - Fatima Cardoso
- grid.421010.60000 0004 0453 9636Breast Unit, Champalimaud Clinical Centre/Champalimaud Foundation, Lisbon, Portugal
| | - Michael Gnant
- grid.22937.3d0000 0000 9259 8492Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Nadia Harbeck
- grid.411095.80000 0004 0477 2585Breast Center, LMU University Hospital, Munich, Germany
| | - Judy King
- grid.437485.90000 0001 0439 3380Royal Free Hospital NHS Foundation Trust, London, UK
| | | | - Frédérique Penault-Llorca
- grid.494717.80000000115480420Centre de Lutte Contre le Cancer Jean Perrin, Imagerie Moléculaire et Stratégies Théranostiques, Université Clermont Auvergne, UMR 1240 INSERM-UCA, Clermont Ferrand, France
| | - Aleix Prat
- grid.10403.360000000091771775Hospital Clínic de Barcelona, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), University of Barcelona, Barcelona, Spain
| |
Collapse
|
10
|
Tutty MA, Holmes S, Prina-Mello A. Cancer Cell Culture: The Basics and Two-Dimensional Cultures. Methods Mol Biol 2023; 2645:3-40. [PMID: 37202610 DOI: 10.1007/978-1-0716-3056-3_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Despite significant advances in investigative and therapeutic methodologies for cancer, 2D cell culture remains an essential and evolving competency in this fast-paced industry. From basic monolayer cultures and functional assays to more recent and ever-advancing cell-based cancer interventions, 2D cell culture plays a crucial role in cancer diagnosis, prognosis, and treatment. Research and development in this field call for a great deal of optimization, while the heterogenous nature of cancer itself demands personalized precision for its intervention. In this way, 2D cell culture is ideal, providing a highly adaptive and responsive platform, where skills can be honed and techniques modified. Furthermore, it is arguably the most efficient, economical, and sustainable methodology available to researchers and clinicians alike.In this chapter, we discuss the history of cell culture and the varying types of cell and cell lines used today, the techniques used to characterize and authenticate them, the applications of 2D cell culture in cancer diagnosis and prognosis, and more recent developments in the area of cell-based cancer interventions and vaccines.
Collapse
Affiliation(s)
- Melissa Anne Tutty
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity College, Dublin, Ireland
| | - Sarah Holmes
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity College, Dublin, Ireland.
| | - Adriele Prina-Mello
- Laboratory of Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity College, Dublin, Ireland
- Nanomedicine and Molecular Imaging Group, Trinity Translational Medicine Institute (TTMI), School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
11
|
Wang Y, Gavan SP, Steinke D, Cheung KL, Chen LC. The impact of age on health utility values for older women with early-stage breast cancer: a systematic review and meta-regression. Health Qual Life Outcomes 2022; 20:169. [PMID: 36564800 PMCID: PMC9789668 DOI: 10.1186/s12955-022-02067-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/07/2022] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION An increasing number of postmenopausal women are diagnosed with breast cancer at an older age (≥ 70 years). There is a lack of synthesised health utility data to support decision-making for managing breast cancer in this older population. This study aimed to identify the availability of, and the subsequent impact of age on, health state utility values (HSUVs) measured by the EQ-5D for older women with early-stage breast cancer. METHOD This systematic review identified EQ-5D (3L or 5L version) HSUVs for postmenopausal women with early-stage breast cancer. Studies were identified from a previous systematic review (inception to 2009) and an electronic database search (Medline and Embase; 2009 to September 2021). Mean HSUVs were summarised by health state. Quality appraisal was performed on studies reporting HSUVs for older ages (≥ 70 years). Multivariable meta-regression assessed the association between HSUVs and age, health state, treatments received, and time of measuring the utility values (greater or less than one year post-treatment). RESULTS Fifty EQ-5D HSUVs were identified from 13 studies. Mean HSUVs decreased as health state worsened: from the stable (mean=0.83) to progression (mean=0.79) and advanced (mean=0.68) states. Two studies reported six HSUVs estimated from the sample of women with a mean age ≥ 70. Meta-regression model fit improved by including age as an independent variable and attenuated the estimated utility decrements associated with worse health states. Utility decrements for the progression and advanced states were -0.052 (95%CI: -0.097, -0.007) and -0.143 (95%CI: -0.264, -0.022) respectively. The breast cancer-specific utility decrement associated with a one-year increase in age was -0.001 (95%CI: -0.004, 0.002). CONCLUSION Relevant and accurate HSUVs are essential to help support decision-making about the most effective and cost-effective ways to manage early-stage breast cancer in older women. Age has a vital role in determining health utility values in this population. This study provides analysts and decision-makers with HSUVs and utility decrements that reflect the disease process in this older population.
Collapse
Affiliation(s)
- Yubo Wang
- grid.5379.80000000121662407Centre for Pharmacoepidemiology and Drug Safety, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Stopford Building, Oxford Road, 1stFloor Stopford Building, Manchester, M13 9PT UK
| | - Sean P. Gavan
- grid.5379.80000000121662407Manchester Centre for Health Economics, Faculty of Biology, Medicine and Health, The University of Manchester, Oxford Road, Manchester, M13 9PL UK
| | - Douglas Steinke
- grid.5379.80000000121662407Centre for Pharmacoepidemiology and Drug Safety, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Stopford Building, Oxford Road, 1stFloor Stopford Building, Manchester, M13 9PT UK
| | - Kwok-Leung Cheung
- grid.4563.40000 0004 1936 8868School of Medicine, University of Nottingham, Royal Derby Hospital Centre, Uttoxeter Road, Derby, DE22 3DT UK
| | - Li-Chia Chen
- grid.5379.80000000121662407Centre for Pharmacoepidemiology and Drug Safety, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Stopford Building, Oxford Road, 1stFloor Stopford Building, Manchester, M13 9PT UK
| |
Collapse
|
12
|
Cree IA, Lokuhetty D, Tan PH. The World Health Organization Classification of Tumors and External Quality Assurance for Immunohistochemistry and Molecular Pathology. Arch Pathol Lab Med 2022; 146:1303-1307. [PMID: 35671156 DOI: 10.5858/arpa.2021-0491-ra] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2021] [Indexed: 11/06/2022]
Abstract
CONTEXT.— This article is based on a talk given by the lead author at the Eigth Annual Princeton Integrated Pathology Symposium: Breast Pathology, on Sunday, April 11, 2021. OBJECTIVE.— To show how the World Health Organization (WHO) Classification of Tumours links to the requirements for quality assurance in breast pathology, including both immunohistochemistry and molecular pathology. DATA SOURCES.— The WHO Classification of Tumours 5th edition Breast Tumours entries formed the basis of the talk, together with guidance published by the International Quality Network for Pathology. CONCLUSIONS.— The WHO Classification of Tumours provides a definitive set of international standards for tumor diagnosis contributed by experts, based on available clinical and research evidence. Techniques used in pathology need internal and external quality assurance to ensure accurate reports for patient management.
Collapse
Affiliation(s)
- Ian A Cree
- From the International Agency for Research on Cancer, World Health Organization, Lyon, France (Cree)
| | - Dilani Lokuhetty
- The Department of Pathology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka (Lokuhetty)
| | - Puay Hoon Tan
- The Division of Pathology, Singapore General Hospital, Singapore (Tan)
| |
Collapse
|
13
|
Rubovszky G, Kocsis J, Boér K, Chilingirova N, Dank M, Kahán Z, Kaidarova D, Kövér E, Krakovská BV, Máhr K, Mriňáková B, Pikó B, Božović-Spasojević I, Horváth Z. Systemic Treatment of Breast Cancer. 1st Central-Eastern European Professional Consensus Statement on Breast Cancer. Pathol Oncol Res 2022; 28:1610383. [PMID: 35898593 PMCID: PMC9311257 DOI: 10.3389/pore.2022.1610383] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/29/2022] [Indexed: 12/11/2022]
Abstract
This text is based on the recommendations accepted by the 4th Hungarian Consensus Conference on Breast Cancer, modified based on the international consultation and conference within the frames of the Central-Eastern European Academy of Oncology. The professional guideline primarily reflects the resolutions and recommendations of the current ESMO, NCCN and ABC5, as well as that of the St. Gallen Consensus Conference statements. The recommendations cover classical prognostic factors and certain multigene tests, which play an important role in therapeutic decision-making. From a didactic point of view, the text first addresses early and then locally advanced breast cancer, followed by locoregionally recurrent and metastatic breast cancer. Within these, we discuss each group according to the available therapeutic options. At the end of the recommendations, we summarize the criteria for treatment in certain rare clinical situations.
Collapse
Affiliation(s)
- Gábor Rubovszky
- Department of Clinical Pharmacology, National Institute of Oncology, Chest and Abdominal Tumours Chemotherapy “B”, Budapest, Hungary,*Correspondence: Gábor Rubovszky,
| | - Judit Kocsis
- Center of Oncoradiology, Bács-Kiskun County Teaching Hospital, Kecskemét, Hungary
| | - Katalin Boér
- Department of Oncology, Szent Margit Hospital, Budapest, Hungary
| | - Nataliya Chilingirova
- Clinic Center of Excellence, Heart and Brain Hospital, Science and Research Institute, Medical University-Pleven, Pleven, Bulgaria
| | - Magdolna Dank
- Oncology Centre, Semmelweis University, Budapest, Hungary
| | | | | | - Erika Kövér
- Institute of Oncotherapy, Faculty of Medicine, University of Pécs, Pécs, Hungary
| | - Bibiana Vertáková Krakovská
- 1st Department of Oncology, Faculty of Medicine, Comenius University, Bratislava, Slovakia,Medical Oncology Department, St. Elisabeth Cancer Institute, Bratislava, Slovakia
| | - Károly Máhr
- Department of Oncology, Szent Rafael Hospital of Zala County, Zalaegerszeg, Hungary
| | - Bela Mriňáková
- 1st Department of Oncology, Faculty of Medicine, Comenius University, Bratislava, Slovakia,Medical Oncology Department, St. Elisabeth Cancer Institute, Bratislava, Slovakia
| | - Béla Pikó
- County Oncology Centre, Pándy Kálmán Hospital of Békés County Council, Gyula, Hungary
| | | | - Zsolt Horváth
- Center of Oncoradiology, Bács-Kiskun County Teaching Hospital, Kecskemét, Hungary
| |
Collapse
|
14
|
Amezcua‑Gálvez J, Lopez‑Garcia C, Villarreal‑Garza C, Lopez‑Rivera V, Canavati‑Marcos M, Santuario‑Facio S, Dono A, Monroig‑Bosque P, Ortiz‑López R, Leal‑Lopez A, Gómez‑macías G. Concordance between Ki‑67 index in invasive breast cancer and molecular signatures: EndoPredict and MammaPrint. Mol Clin Oncol 2022; 17:132. [PMID: 35949891 PMCID: PMC9353786 DOI: 10.3892/mco.2022.2565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/17/2022] [Indexed: 12/24/2022] Open
Abstract
Identifying patients with hormone receptor-positive (HR+) early invasive breast cancer (EIBC) who benefit from adjuvant chemotherapy has improved with molecular signature tests. However, due to high cost and limited availability, alternative tests are used. The present study sought to evaluate the performance of the proliferation marker Ki-67 to identify these patients and explore its association with molecular signatures and risk stratification markers. From the San José TecSalud Hospital in Monterrey México, patients with HR+ EIBC as tested with EndoPredict or MammaPrint and Ki-67 index were identified. They were categorized into two groups: Group 1 (June 2016-August 2018) was evaluated using EndoPredict and Group 2 (June 2016-August 2018) with MammaPrint. A ≥20% Ki67 index cutoff was utilized to identify highly proliferative EIBC and an area under the receiver-operating characteristic curve and κ concordance were utilized to evaluate the performance of Ki-67 index compared to molecular signature tests. In the EndoPredict group, 54/96 patients were considered high-risk based on their EPclin score, while 57/96 patients had Ki-67 index ≥20%. However, there was no significant overall concordance between them (59.37%, κ=0.168, P=0.09), while the given risk of distant recurrence given in percentage by EPclin had a positive association with the Ki67 index (P=0.04). In the MammaPrint group, 21/70 patients were considered high-risk and 36/70 patients presented with a Ki-67 index ≥20% with a significant overall concordance (67.14%, κ=0.35, P<0.001). In addition, high Ki-67 index was associated with the Nottingham histological grade in both groups. In conclusion, there was a concordance between Ki-67 and MammaPrint risk stratification of HR+ EIBC and no concordance with the EndoPredict molecular signature, but a positive association with the given percentage of recurrence and the median Ki-67 index as the cutoff at our center. Cost-effectiveness analyses of these tests in developing countries are required; until then, the use of Ki-67 appears reasonable to aid clinical decisions, together with the other established clinicopathological variables.
Collapse
Affiliation(s)
- Jesús Amezcua‑Gálvez
- Department of Pathology, Tecnologico de Monterrey, Hospital San José, Av. Morones Prieto Poniente 3000 Poniente, Los Doctores, Monterrey, NL 64710, México
| | - Carlos Lopez‑Garcia
- Department of Pathology, Tecnologico de Monterrey, Hospital San José, Av. Morones Prieto Poniente 3000 Poniente, Los Doctores, Monterrey, NL 64710, México
| | - Cynthia Villarreal‑Garza
- Breast Cancer Center, Tecnologico de Monterrey, Hospital Zambrano Hellion, Real San Agustín, San Pedro Garza García, NL 66278, México
| | - Victor Lopez‑Rivera
- Department of Neurology, UTHealth McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Mauricio Canavati‑Marcos
- Breast Cancer Center, Tecnologico de Monterrey, Hospital Zambrano Hellion, Real San Agustín, San Pedro Garza García, NL 66278, México
| | - Sandra Santuario‑Facio
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Av Morones Prieto, 3000 Poniente, Los Doctores, Monterrey, Nuevo León, 64710, México
| | - Antonio Dono
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, TX 77030, USA
| | - Paloma Monroig‑Bosque
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Rocío Ortiz‑López
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Av Morones Prieto, 3000 Poniente, Los Doctores, Monterrey, Nuevo León, 64710, México
| | - Andrea Leal‑Lopez
- Breast Cancer Center, Tecnologico de Monterrey, Hospital Zambrano Hellion, Real San Agustín, San Pedro Garza García, NL 66278, México
| | - Gabriela Gómez‑macías
- Department of Pathology, Tecnologico de Monterrey, Hospital San José, Av. Morones Prieto Poniente 3000 Poniente, Los Doctores, Monterrey, NL 64710, México
| |
Collapse
|
15
|
Jank P, Lindner JL, Lehmann A, Pfitzner BM, Blohmer JU, Horst D, Kronenwett R, Denkert C, Schmitt WD. Comparison of risk assessment in 1652 early ER positive, HER2 negative breast cancer in a real-world data set: classical pathological parameters vs. 12-gene molecular assay (EndoPredict). Breast Cancer Res Treat 2022; 191:327-333. [PMID: 34783927 PMCID: PMC8763835 DOI: 10.1007/s10549-021-06415-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/08/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Risk assessment on the molecular level is important in predictive pathology to determine the risk of metastatic disease for ERpos, HER2neg breast cancer. The gene expression test EndoPredict (EP) was trained and validated for prediction of a 10-year risk of distant recurrence to support therapy decisions regarding endocrine therapy alone or in combination with chemotherapy. The EP test provides the 12-gene Molecular Score (MS) and the EPclin-Score (EPclin), which combines the molecular score with tumor size and nodal status. In this project we investigated the correlation of 12-gene MS and EPclin scores with classical pathological markers. METHODS EndoPredict-based gene expression profiling was performed prospectively in a total of 1652 patients between 2017 and 2020. We investigated tumor grading and Ki67 cut-offs of 20% for binary classification as well as 10% and 30% for three classes (low, intermediate, high), based on national and international guidelines. RESULTS 410 (24.8%) of 1652 patients were classified as 12-gene MS low risk and 626 (37.9%) as EPclin low risk. We found significant positive associations between 12-gene MS and grading (p < 0.001), EPclin and grading (p = 0.001), 12-gene MS and Ki67 (p < 0.001), and EPclin and Ki67 (p < 0.001). However, clinically relevant differences between EP test results, Ki67 and tumor grading were observed. For example, 118 (26.3%) of 449 patients with Ki67 > 20% were classified as low risk by EPclin. Same differences were seen comparing EP test results and tumor grading. CONCLUSION In this study we could show that EP risk scores are distributed differentially among Ki67 expression groups, especially in Ki67 low and high tumors with a substantial proportion of patients with EPclin high risk results in Ki67 low tumors and vice versa. This suggests that classical pathological parameters and gene expression parameters are not interchangeable, but should be used in combination for risk assessment.
Collapse
Affiliation(s)
- Paul Jank
- Institute of Pathology, Philipps-University Marburg and University Hospital Marburg (UKGM)-Universitätsklinikum Marburg, Baldingerstraße, 35043, Marburg, Germany
| | - Judith Lea Lindner
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | - Annika Lehmann
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | | | - Jens-Uwe Blohmer
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Department of Gynecology and Obstetrics, Charitéplatz 1, 10117, Berlin, Germany
| | - David Horst
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| | | | - Carsten Denkert
- Institute of Pathology, Philipps-University Marburg and University Hospital Marburg (UKGM)-Universitätsklinikum Marburg, Baldingerstraße, 35043, Marburg, Germany.
| | - Wolfgang Daniel Schmitt
- Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Pathology, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
16
|
Scarborough JA, Scott JG. Translation of Precision Medicine Research Into Biomarker-Informed Care in Radiation Oncology. Semin Radiat Oncol 2022; 32:42-53. [PMID: 34861995 PMCID: PMC8667861 DOI: 10.1016/j.semradonc.2021.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The reach of personalized medicine in radiation oncology has expanded greatly over the past few decades as technical precision has improved the delivery of radiation to each patient's unique anatomy. Yet, the consideration of biological heterogeneity between patients has largely not been translated to clinical care. There are innumerable promising advancements in the discovery and validation of biomarkers, which could be used to alter radiation therapy directly or indirectly. Directly, biomarker-informed care may alter treatment dose or identify patients who would benefit most from radiation therapy and who could safely avoid more aggressive care. Indirectly, a variety of biomarkers could assist with choosing the best radiosensitizing chemotherapies. The translation of these advancements into clinical practice will bring radiation oncology even further into the era of precision medicine, treating patients according to their unique anatomical and biological differences.
Collapse
Affiliation(s)
- Jessica A Scarborough
- Translational Hematology and Oncology Research Department, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland,OH; Systems Biology and Bioinformatics Program, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Jacob G Scott
- Translational Hematology and Oncology Research Department, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland,OH; Radiation Oncology Department, Taussig Cancer Institute, Cleveland Clinic Foundation, 10201 Carnegie Ave, Cleveland, OH.
| |
Collapse
|
17
|
Maniez P, Osada M, Reix N, Mathelin C. [uPA/PAI-1 and EPClin®: Comparison of their impact on the management of intermediate-prognosis breast cancers]. ACTA ACUST UNITED AC 2021; 50:298-306. [PMID: 34626849 DOI: 10.1016/j.gofs.2021.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The uPA/PAI-1 assay and the EPClin® test are useful tools that add to clinico-anatomical characteristics to determine the indication of adjuvant chemotherapy in case of intermediate-prognosis invasive breast cancer. The principal purpose of our study was to analyze the concordance of uPA/PAI-1 and EPClin® in classification of patients into two groups: low and high risk of relapse. METHODS We prospectively included 63 patients treated for intermediate-prognosis invasive breast cancer. All of these patients received a uPA/PAI-1 assay and an EPClin® test. RESULTS The uPA/PAI-1 assay and EPClin® test were consistent for 56.2% and inconsistent for 43.8%. In the event of a discrepancy, the treatment decision was based in 95.2% of patients on the EPClin® test result. In total, 38 patients were selected for adjuvant chemotherapy after achievement of the two tests. The mean time to report results after surgery was 9 days for the uPA/PAI-1 assay and 35 days for the EPClin® test. No cases of recurrence or death were found, with an average follow-up of 32 months. CONCLUSION The EPClin® test resulted in more chemotherapy prescriptions than indicated by uPA/PAI-1. However, we can't conclude to the superiority of one of these two tests, survival data and the effectiveness of our study being insufficient. In general, studies comparing different signatures useful to the therapeutic decision of intermediate prognosis breast cancers should be encouraged.
Collapse
Affiliation(s)
- P Maniez
- Hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France; Service de chirurgie, Institut de cancérologie Strasbourg Europe (ICANS), 17, rue Albert-Calmette, 67200 Strasbourg, France.
| | - M Osada
- Hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France; Service de chirurgie, Institut de cancérologie Strasbourg Europe (ICANS), 17, rue Albert-Calmette, 67200 Strasbourg, France
| | - N Reix
- ICube UMR 7357, université de Strasbourg/CNRS, Fédération de médecine translationnelle de Strasbourg (FMTS), Strasbourg, France; Laboratoire de biochimie et biologie moléculaire, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France
| | - C Mathelin
- Hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France; Service de chirurgie, Institut de cancérologie Strasbourg Europe (ICANS), 17, rue Albert-Calmette, 67200 Strasbourg, France; CNRS UMR7104 Inserm U964, Institut de génétique et de biologie moléculaire et cellulaire (IGBMC), 1, rue Laurent-Fries, 67400 Illkirch-Graffenstaden, France
| |
Collapse
|
18
|
Salgado R, Peg V, Rüschoff J, Vincent-Salomon A, Castellano I, Perner S, Van de Vijver K, Quinn CM, Varga Z. Gene expression signatures for tailoring adjuvant chemotherapy of luminal breast cancer: the pathologists' perspective. Ann Oncol 2021; 32:1316-1321. [PMID: 34461263 DOI: 10.1016/j.annonc.2021.08.1993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/04/2021] [Accepted: 08/22/2021] [Indexed: 10/20/2022] Open
Affiliation(s)
- R Salgado
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium; Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia.
| | - V Peg
- Universidad Autónoma de Barcelona, Barcelona, Spain; Department of Pathology, Vall D'Hebron University Hospital, Barcelona, Spain; Spanish Biomedical Research Network Centre in Oncology (CIBERONC), Madrid, Spain
| | - J Rüschoff
- Targos Molecular Pathology GmbH and Institute of Pathology Nordhessen, Kassel, Germany
| | - A Vincent-Salomon
- Department of Pathology and Department of Diagnostic and Theranostic Medicine, Institut Curie, PSL Research University, Paris, France
| | - I Castellano
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - S Perner
- Institute of Pathology, University Hospital of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany; Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - K Van de Vijver
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - C M Quinn
- Department of Pathology, St. Vincent's University Hospital, Dublin and University College Dublin, Dublin, Ireland
| | - Z Varga
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
Prognostic and predictive markers for adjuvant therapy. Curr Opin Obstet Gynecol 2021; 32:100-105. [PMID: 31833940 DOI: 10.1097/gco.0000000000000594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW To avoid both overtreatment and undertreatment accurate risk assessment is mandatory. The present review gives an overview of recently published articles covering prognostic and predictive factors for adjuvant therapy in early breast cancer. RECENT FINDINGS Gene expression signatures enhance prognostic accuracy with a high level of evidence. These signatures can be further improved by incorporating traditional pathological factors like tumor size. Newer genomic techniques like next-generation sequencing lead to a deeper understanding of the relationship between somatic mutations and prognosis or prediction of therapeutic efficacy. Furthermore, circulating tumor cells, and circulating cell-free or tumor DNA can lead to a better estimation of the risk of recurrence in early breast cancer. In addition, recent results underscore the prognostic and predictive importance of tumor-infiltrating lymphocytes and subtyping of immune cell infiltrates especially in triple-negative breast cancer. SUMMARY The current review highlights recent studies improving prognostication and prediction of therapeutic efficacy in early breast cancer. These advances should lead to a better risk stratification and thereby to an improved tailoring of therapies.
Collapse
|
20
|
Li X, Sun H, Liu Q, Liu Y, Hou Y, Jin W. A pharmacophore-based classification better predicts the outcomes of HER2-negative breast cancer patients receiving the anthracycline- and/or taxane-based neoadjuvant chemotherapy. Cancer Med 2021; 10:4658-4674. [PMID: 34076352 PMCID: PMC8267145 DOI: 10.1002/cam4.4022] [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: 11/09/2020] [Revised: 04/13/2021] [Accepted: 05/05/2021] [Indexed: 12/31/2022] Open
Abstract
AIMS Prognosis of patients for human epidermal growth factor receptor 2 (HER2)-negative breast cancer post neoadjuvant chemotherapy is not well understood. The aim of this study was to develop a novel pharmacophore-based signature to better classify and predict the risk of HER2-negative patients after anthracycline-and/or taxane-based neoadjuvant chemotherapy (NACT). MAIN METHODS Anthracycline and taxane pharmacophore-based genes were obtained from PharmMapper. Drug-targeted genes (DTG) related clinical and bioinformatic analyses were undertaken in four GEO datasets. KEY FINDINGS We used 12 genes from the pharmacophore to develop a DTG score (DTG-S). The DTG-S classification exhibited significant prognostic ability with respect to disease free survival (DFS) for HER2-negative patients who receive at least one type of neoadjuvant chemotherapy that included anthracycline and/or taxane. DTG-S associated with a high predictive ability for pathological complete response (pCR) as well as for prognosis of breast cancer. Using the DTG-S classification in other prediction models may improve the reclassification accuracy for DFS. Combining the DTG-S with other clinicopathological factors may further improve its predictive ability of patients' outcomes. Gene ontology and KEGG pathway analysis showed that the biological processes of DTG-S high group were associated with the cell cycle, cell migration, and cell signal transduction pathways. Targeted drug analysis shows that some CDK inhibitors and PI3K-AKT pathway inhibitors may be useful for high DTG-S patients. SIGNIFICANCE The DTG-S classification adds prognostic and predictive information to classical parameters for HER2-negative patients who receive anthracycline-and/or taxane-based NACT, which could improve the patients' risk stratification and may help guide adjuvant treatment.
Collapse
Affiliation(s)
- Xuan Li
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hefen Sun
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiqi Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yang Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yifeng Hou
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Jin
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Vatish J, Wilkinson B, Al-Ishaq Z, Pujji O, Isgar B, Vidya R, Matey P, Sircar T, Mylvaganam S. The use of genomic assays reduces rates of chemotherapy: a single-institution experience. Ir J Med Sci 2021; 191:687-690. [PMID: 33993406 DOI: 10.1007/s11845-021-02650-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The National Institute for Clinical Excellence recommends the use of tumour profiling tests to guide adjuvant chemotherapy in breast cancer. The Oncotype DX™ score (Genomic Health) has superseded more traditional tools such as PREDICT in appropriate patients (ER + ve, HER2-ve, lymph node negative and with a Nottingham Prognostic Index [NPI] ≥ 3.4). The aim of this study was to see whether the introduction of Oncotype DX within our institution resulted in an overall reduction in rates of chemotherapy. METHOD Data was collected retrospectively using the Somerset Cancer Register, Pathology department databases and the institution's own online medical records system. Two groups were compared: (1) pre-oncotype (Jan 2012-Dec 2014) and (2) post-oncotype (Jan 2016-July 2018). RESULTS During the pre-oncotype period, 28/82 (34%) patients who would have been eligible for testing (patients who were ER + ve, HER2-ve, and a NPI ≥ 3.4) received chemotherapy compared to 34/135 (25%) who were sent for oncotype during the second study period (p = 0.157). For grade 3 cancers, and those aged under 50, the results were more marked: grade 3 pre-oncotype 23/43 (53%), post-oncotype 29/76 (38%) (p = 0.101), aged under 50 pre-oncotype 8/15 (53%), post-oncotype 10/31 (32%) (p = 0.197). CONCLUSION Within our institution, overall rates of chemotherapy have reduced since the introduction of Oncotype DX with the results more marked in subgroups of traditional indicators of tumour aggression. As genomic assays provide a more accurate prediction of the benefit of chemotherapy, its overall reduction has potential cost saving implications as well as reducing risk in patients who will derive little benefit.
Collapse
Affiliation(s)
- Jamie Vatish
- Corporate Services Centre, Royal Wolverhampton NHS Trust 12, New Cross Hospital, Wolverhampton, WV10 0QP, UK.
| | - Ben Wilkinson
- Corporate Services Centre, Royal Wolverhampton NHS Trust 12, New Cross Hospital, Wolverhampton, WV10 0QP, UK
| | - Zaid Al-Ishaq
- Corporate Services Centre, Royal Wolverhampton NHS Trust 12, New Cross Hospital, Wolverhampton, WV10 0QP, UK
| | - Ojas Pujji
- Corporate Services Centre, Royal Wolverhampton NHS Trust 12, New Cross Hospital, Wolverhampton, WV10 0QP, UK
| | - Brian Isgar
- Corporate Services Centre, Royal Wolverhampton NHS Trust 12, New Cross Hospital, Wolverhampton, WV10 0QP, UK
| | - Raghavan Vidya
- Corporate Services Centre, Royal Wolverhampton NHS Trust 12, New Cross Hospital, Wolverhampton, WV10 0QP, UK
| | - Pilar Matey
- Corporate Services Centre, Royal Wolverhampton NHS Trust 12, New Cross Hospital, Wolverhampton, WV10 0QP, UK
| | - Tapan Sircar
- Corporate Services Centre, Royal Wolverhampton NHS Trust 12, New Cross Hospital, Wolverhampton, WV10 0QP, UK
| | - Senthuran Mylvaganam
- Corporate Services Centre, Royal Wolverhampton NHS Trust 12, New Cross Hospital, Wolverhampton, WV10 0QP, UK
| |
Collapse
|
22
|
Real-world analysis of clinical and economic impact of 21-gene recurrence score (RS) testing in early-stage breast cancer (ESBC) in Ireland. Breast Cancer Res Treat 2021; 188:789-798. [PMID: 33835293 DOI: 10.1007/s10549-021-06211-w] [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: 11/18/2020] [Accepted: 03/23/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE Results from TAILOR-X suggest that up to 70% of hormone receptor-positive (HR+) node-negative (N0) ESBC patients (pts) may avoid chemotherapy (CT) with RS ≤ 25. We assess clinical and economic impacts of RS testing on treatment using real-world data. METHODS From October 2011 to February 2019, a retrospective, cross-sectional observational study was conducted of HR+ N0 ESBC pts who had RS testing in Ireland. Pts were classified low risk (RS ≤ 25) and high risk (RS > 25). Clinical risk was calculated. Data were collected via electronic patient records. Cost data were supplied by the National Healthcare Pricing Regulatory Authority. RESULTS 963 pts. Mean age is 56 years. Mean tumour size is 1.7 cm. 114 (11.8%), 635 (66%), 211 (22%), 3 (0.2%) pts had G1, G2, G3 and unknown G, respectively. 796 pts (82.8%) low RS, 159 (16.5%) high RS and 8 pts (0.7%) unknown RS. 263 pts (26%) were aged ≤ 50 at diagnosis; 117 (45%) had RS 0-15, 63 (24.5%) 16-20, 39 (15.3%) 21-25 and 40 (15.2%) RS 26-100. 4 pts (1.5%) had unknown RS. Post-RS testing, 602 pts (62.5%) had a change in CT decision; 593 changed to hormone therapy (HT) alone. In total, 262 pts received CT. Of pts receiving CT; 138 (53%) had RS > 25, 124 (47%) had RS ≤ 25. Of pts aged ≤ 50, 153 (58%) had high clinical risk, of whom 28 had RS 16-20. Assay use achieved a 62.5% change in treatment with 73% of pts avoiding CT. This resulted in savings of €4 million in treatment costs. Deducting assay costs, savings of €1.9 million were achieved. CONCLUSION Over the 8 years of the study, a 62.5% reduction in CT use was achieved with savings of over €1,900,000.
Collapse
|
23
|
Gild ML, Tsang VHM, Clifton-Bligh RJ, Robinson BG. Multikinase inhibitors in thyroid cancer: timing of targeted therapy. Nat Rev Endocrinol 2021; 17:225-234. [PMID: 33603220 DOI: 10.1038/s41574-020-00465-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/18/2020] [Indexed: 02/06/2023]
Abstract
In the 9 years since the publication of our 2011 review of targeted treatment of thyroid cancer with multikinase inhibitors, much has changed in the landscape of this heterogeneous disease. New multikinase and selective inhibitor treatments for medullary thyroid cancer, radioiodine-refractory thyroid cancer and anaplastic thyroid cancer have completed trials and improved progression-free survival. Many physicians are concerned by dose-limiting adverse effects of these drugs and are wary to begin treatment in patients who are systemically well but have marked disease burden, which makes the timing of treatment initiation challenging. Published mechanistic data on tyrosine kinase inhibitors (TKIs) have helped guide our understanding of how to dose effectively with these drugs. A major goal in TKI therapy is to optimize inhibition of oncogenic kinase drivers while maintaining patient quality of life. Real-world data have now been published on how TKIs have fared outside the clinical trial environment. In this Review, we provide a summary of published data on the efficacy of TKIs in clinical practice, to provide clinicians with a more realistic view of how their patients will manage and respond to TKI therapy. Furthermore, we review the data on mechanisms of inhibition, outcomes and adverse effects of TKIs and provide an update on targeted treatment of thyroid cancer, focusing on optimizing the timing of treatment initiation.
Collapse
Affiliation(s)
- Matti L Gild
- Department of Endocrinology and Diabetes, Royal North Shore Hospital, Sydney, NSW, Australia.
- Cancer Genetics, Kolling Institute of Medical Research, Sydney, NSW, Australia.
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.
| | - Venessa H M Tsang
- Department of Endocrinology and Diabetes, Royal North Shore Hospital, Sydney, NSW, Australia
- Cancer Genetics, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Roderick J Clifton-Bligh
- Department of Endocrinology and Diabetes, Royal North Shore Hospital, Sydney, NSW, Australia
- Cancer Genetics, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Bruce G Robinson
- Department of Endocrinology and Diabetes, Royal North Shore Hospital, Sydney, NSW, Australia
- Cancer Genetics, Kolling Institute of Medical Research, Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
24
|
Thiruthaneeswaran N, Bibby BAS, Yang L, Hoskin PJ, Bristow RG, Choudhury A, West C. Lost in application: Measuring hypoxia for radiotherapy optimisation. Eur J Cancer 2021; 148:260-276. [PMID: 33756422 DOI: 10.1016/j.ejca.2021.01.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/21/2021] [Accepted: 01/28/2021] [Indexed: 12/15/2022]
Abstract
The history of radiotherapy is intertwined with research on hypoxia. There is level 1a evidence that giving hypoxia-targeting treatments with radiotherapy improves locoregional control and survival without compromising late side-effects. Despite coming in and out of vogue over decades, there is now an established role for hypoxia in driving molecular alterations promoting tumour progression and metastases. While tumour genomic complexity and immune profiling offer promise, there is a stronger evidence base for personalising radiotherapy based on hypoxia status. Despite this, there is only one phase III trial targeting hypoxia modification with full transcriptomic data available. There are no biomarkers in routine use for patients undergoing radiotherapy to aid management decisions, and a roadmap is needed to ensure consistency and provide a benchmark for progression to application. Gene expression signatures address past limitations of hypoxia biomarkers and could progress biologically optimised radiotherapy. Here, we review recent developments in generating hypoxia gene expression signatures and highlight progress addressing the challenges that must be overcome to pave the way for their clinical application.
Collapse
Affiliation(s)
- Niluja Thiruthaneeswaran
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.
| | - Becky A S Bibby
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Lingjang Yang
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Peter J Hoskin
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; Mount Vernon Cancer Centre, Northwood, UK
| | - Robert G Bristow
- Division of Cancer Sciences, The University of Manchester, Manchester, UK; CRUK Manchester Institute and Manchester Cancer Research Centre, Manchester, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, The University of Manchester, Christie Hospital NHS Foundation Trust, Manchester, UK
| | - Catharine West
- Division of Cancer Sciences, The University of Manchester, Christie Hospital NHS Foundation Trust, Manchester, UK
| |
Collapse
|
25
|
Giorgi Rossi P, Lebeau A, Canelo-Aybar C, Saz-Parkinson Z, Quinn C, Langendam M, Mcgarrigle H, Warman S, Rigau D, Alonso-Coello P, Broeders M, Graewingholt A, Posso M, Duffy S, Schünemann HJ. Recommendations from the European Commission Initiative on Breast Cancer for multigene testing to guide the use of adjuvant chemotherapy in patients with early breast cancer, hormone receptor positive, HER-2 negative. Br J Cancer 2021; 124:1503-1512. [PMID: 33597715 PMCID: PMC8076250 DOI: 10.1038/s41416-020-01247-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/10/2020] [Accepted: 12/17/2020] [Indexed: 12/12/2022] Open
Abstract
Background Predicting the risk of recurrence and response to chemotherapy in women with early breast cancer is crucial to optimise adjuvant treatment. Despite the common practice of using multigene tests to predict recurrence, existing recommendations are inconsistent. Our aim was to formulate healthcare recommendations for the question “Should multigene tests be used in women who have early invasive breast cancer, hormone receptor-positive, HER2-negative, to guide the use of adjuvant chemotherapy?” Methods The European Commission Initiative on Breast Cancer (ECIBC) Guidelines Development Group (GDG), a multidisciplinary guideline panel including experts and three patients, developed recommendations informed by systematic reviews of the evidence. Grading of Recommendations Assessment, Development and Evaluation (GRADE) Evidence to Decision frameworks were used. Four multigene tests were evaluated: the 21-gene recurrence score (21-RS), the 70-gene signature (70-GS), the PAM50 risk of recurrence score (PAM50-RORS), and the 12-gene molecular score (12-MS). Results Five studies (2 marker-based design RCTs, two treatment interaction design RCTs and 1 pooled individual data analysis from observational studies) were included; no eligible studies on PAM50-RORS or 12-MS were identified and the GDG did not formulate recommendations for these tests. Conclusions The ECIBC GDG suggests the use of the 21-RS for lymph node-negative women (conditional recommendation, very low certainty of evidence), recognising that benefits are probably larger in women at high risk of recurrence based on clinical characteristics. The ECIBC GDG suggests the use of the 70-GS for women at high clinical risk (conditional recommendation, low certainty of evidence), and recommends not using 70-GS in women at low clinical risk (strong recommendation, low certainty of evidence).
Collapse
Affiliation(s)
- Paolo Giorgi Rossi
- Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Annette Lebeau
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carlos Canelo-Aybar
- Iberoamerican Cochrane Center, Biomedical Research Institute (IIB Sant Pau-CIBERESP), Barcelona, Spain.,Department of Paediatrics, Obstetrics and Gynaecology, Preventive Medicine, and Public Health, PhD Programme in Methodology of Biomedical Research and Public Health, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Zuleika Saz-Parkinson
- European Commission, Joint Research Centre (JRC), Ispra, Italy. .,Instituto de Salud Carlos III, Health Technology Assessment Agency, Avenida Monforte de Lemos 5, Madrid, Spain.
| | - Cecily Quinn
- St. Vincent's University Hospital, Dublin, Ireland
| | - Miranda Langendam
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health Institute, Amsterdam, The Netherlands
| | | | - Sue Warman
- Havyatt Lodge, Havyatt Road, Langford, North Somerset, UK
| | - David Rigau
- Iberoamerican Cochrane Center, Biomedical Research Institute (IIB Sant Pau-CIBERESP), Barcelona, Spain
| | - Pablo Alonso-Coello
- Iberoamerican Cochrane Center, Biomedical Research Institute (IIB Sant Pau-CIBERESP), Barcelona, Spain
| | - Mireille Broeders
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, the Netherlands.,Dutch Expert Centre for Screening, Nijmegen, the Netherlands
| | | | - Margarita Posso
- Iberoamerican Cochrane Center, Biomedical Research Institute (IIB Sant Pau-CIBERESP), Barcelona, Spain.,Department of Epidemiology and Evaluation, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain.,Research Network on Health Services in Chronic Diseases (REDISSEC), Barcelona, Spain
| | - Stephen Duffy
- Centre for Cancer Prevention, Queen Mary University of London, Charterhouse Square, London, UK
| | - Holger J Schünemann
- Michael G. DeGroote Cochrane Canada and McGRADE Centres; Department of Health Research Methods, Evidence and Impact, McMaster University Health Sciences Centre, Hamilton, Ontario, Canada
| | | |
Collapse
|
26
|
Buus R, Szijgyarto Z, Schuster EF, Xiao H, Haynes BP, Sestak I, Cuzick J, Paré L, Seguí E, Chic N, Prat A, Dowsett M, Cheang MCU. Development and validation for research assessment of Oncotype DX® Breast Recurrence Score, EndoPredict® and Prosigna®. NPJ Breast Cancer 2021; 7:15. [PMID: 33579961 PMCID: PMC7881187 DOI: 10.1038/s41523-021-00216-w] [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: 06/03/2020] [Accepted: 12/17/2020] [Indexed: 12/22/2022] Open
Abstract
Multi-gene prognostic signatures including the Oncotype® DX Recurrence Score (RS), EndoPredict® (EP) and Prosigna® (Risk Of Recurrence, ROR) are widely used to predict the likelihood of distant recurrence in patients with oestrogen-receptor-positive (ER+), HER2-negative breast cancer. Here, we describe the development and validation of methods to recapitulate RS, EP and ROR scores from NanoString expression data. RNA was available from 107 tumours from postmenopausal women with early-stage, ER+, HER2- breast cancer from the translational Arimidex, Tamoxifen, Alone or in Combination study (TransATAC) where previously these signatures had been assessed with commercial methodology. Gene expression was measured using NanoString nCounter. For RS and EP, conversion factors to adjust for cross-platform variation were estimated using linear regression. For ROR, the steps to perform subgroup-specific normalisation of the gene expression data and calibration factors to calculate the 46-gene ROR score were assessed and verified. Training with bootstrapping (n = 59) was followed by validation (n = 48) using adjusted, research use only (RUO) NanoString-based algorithms. In the validation set, there was excellent concordance between the RUO scores and their commercial counterparts (rc(RS) = 0.96, 95% CI 0.93-0.97 with level of agreement (LoA) of -7.69 to 8.12; rc(EP) = 0.97, 95% CI 0.96-0.98 with LoA of -0.64 to 1.26 and rc(ROR) = 0.97 (95% CI 0.94-0.98) with LoA of -8.65 to 10.54). There was also a strong agreement in risk stratification: (RS: κ = 0.86, p < 0.0001; EP: κ = 0.87, p < 0.0001; ROR: κ = 0.92, p < 0.001). In conclusion, the calibrated algorithms recapitulate the commercial RS and EP scores on individual biopsies and ROR scores on samples based on subgroup-centreing method using NanoString expression data.
Collapse
Affiliation(s)
- Richard Buus
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - Zsolt Szijgyarto
- Clinical Trials and Statistics Unit (ICR-CTSU), Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - Eugene F Schuster
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - Hui Xiao
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - Ben P Haynes
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | | | | | - Laia Paré
- Department of Medical Oncology, Hospital Clinic, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
| | - Elia Seguí
- Department of Medical Oncology, Hospital Clinic, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
| | - Nuria Chic
- Department of Medical Oncology, Hospital Clinic, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
| | - Aleix Prat
- Department of Medical Oncology, Hospital Clinic, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
| | - Mitch Dowsett
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - Maggie Chon U Cheang
- Clinical Trials and Statistics Unit (ICR-CTSU), Division of Clinical Studies, The Institute of Cancer Research, London, UK.
| |
Collapse
|
27
|
A clinical calculator to predict disease outcomes in women with triple-negative breast cancer. Breast Cancer Res Treat 2021; 185:557-566. [PMID: 33389409 DOI: 10.1007/s10549-020-06030-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/16/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, characterized by substantial risks of early disease recurrence and mortality. We constructed and validated clinical calculators for predicting recurrence-free survival (RFS) and overall survival (OS) for TNBC. METHODS Data from 605 women with centrally confirmed TNBC who underwent primary breast cancer surgery at Mayo Clinic during 1985-2012 were used to train risk models. Variables included age, menopausal status, tumor size, nodal status, Nottingham grade, surgery type, adjuvant radiation therapy, adjuvant chemotherapy, Ki67, stromal tumor-infiltrating lymphocytes (sTIL) score, and neutrophil-to-lymphocyte ratio (NLR). Final models were internally validated for calibration and discrimination using ten-fold cross-validation and compared with their base-model counterparts which include only tumor size and nodal status. Independent external validation was performed using data from 478 patients diagnosed with stage II/III invasive TNBC during 1986-1992 in the British Columbia Breast Cancer Outcomes Unit database. RESULTS Final RFS and OS models were well calibrated and associated with C-indices of 0.72 and 0.73, as compared with 0.64 and 0.62 of the base models (p < 0.001). In external validation, the discriminant ability of the final models was comparable to the base models (C-index: 0.59-0.61). The RFS model demonstrated greater accuracy than the base model both overall and within patient subgroups, but the advantages of the OS model were less profound. CONCLUSIONS This TNBC clinical calculator can be used to predict patient outcomes and may aid physician's communication with TNBC patients regarding their long-term disease outlook and planning treatment strategies.
Collapse
|
28
|
Bernhardt SM, Dasari P, Walsh D, Raymond W, Hull ML, Townsend AR, Price TJ, Ingman WV. The menstrual cycle is an under-appreciated factor in premenopausal breast cancer diagnosis and treatment. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.coemr.2020.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
29
|
Gordon-Craig S, Parks RM, Cheung KL. The Potential Use of Tumour-Based Prognostic and Predictive Tools in Older Women with Primary Breast Cancer: A Narrative Review. Oncol Ther 2020; 8:231-250. [PMID: 32700048 PMCID: PMC7366554 DOI: 10.1007/s40487-020-00123-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Indexed: 01/09/2023] Open
Abstract
A move is under way towards personalised cancer treatment, where tumour biology of an individual patient is examined to give unique predictive and prognostic information. This is extremely important in the setting of older women, who have treatment-specific goals which may differ from their younger counterparts, and may include conservation of quality of life rather than curative intent of treatment. One method employed to assist with this is the use of tumour-based prognostic and predictive tools. This article explores six of the most common tumour-based tools currently available on the market: MammaPrint, Oncotype DX, Mammostrat, Prosigna, EndoPredict, IHC4. The article discusses the creation and validation of these tools, their use and validation in older women, and future directions in the field. With the exception of Oncotype Dx, which has also been licensed for prediction of response from adjuvant chemotherapy, these tools have been licensed for use as prognostic tools only, mainly in the setting of adjuvant therapy following surgery. The evidence base for use in older women is strongest for Mammostrat and PAM50, although overall the evidence is much weaker than that in younger women. Where older women have been included in validation studies, this is often in small numbers, or the exact proportion of older women is unknown. In practice, all six of the tools are recommended to be utilised on surgical excision specimens, as well as in core needle biopsy (CNB) specimens in all of the tools except Mammostrat. This is extremely important in the setting of older women, of whom a large proportion do not undergo surgery. The suggested nature of the sample is formalin-fixed paraffin-embedded in all the tools except MammaPrint, which can also be performed on fresh-frozen samples. Future development of prognostic tools in older women with breast cancer should focus on treatment dilemmas specific to this population. This includes the decision of primary treatment between surgery or endocrine therapy and decisions regarding adjuvant therapy, in particular, chemotherapy.
Collapse
Affiliation(s)
- Sophie Gordon-Craig
- Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Ruth M Parks
- Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Kwok-Leung Cheung
- Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham, Nottingham, UK.
| |
Collapse
|
30
|
Jin T, Zhang Y, Zhang T. MiR-524-5p Suppresses Migration, Invasion, and EMT Progression in Breast Cancer Cells Through Targeting FSTL1. Cancer Biother Radiopharm 2020; 35:789-801. [PMID: 32298609 DOI: 10.1089/cbr.2019.3046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: The effects of miR-524-5p on breast cancer (BC) have not been investigated, though studies show that miR-524-5p has an anticancer function. Thus, this study investigated the effects of miR-524-5p on BC cells and its potential molecular mechanism. Materials and Methods: The expression of miR-524-5p from the collected BC samples was determined. Cell counting kit-8 (CCK-8) assay was performed to examine the effect of miR-524-5p on BC cells viability. The target for miR-524-5p was predicted by bioinformatics and further verified by luciferase assay. Wound healing assay and transwell assay were performed to determine cell migration and invasion of BC cells. The expressions of Follistatin-like 1 (FSTL1) and related proteins in epithelial-mesenchymal transition (EMT) were detected by Western blotting and quantitative real-time polymerase chain reaction. Results: MiR-524-5p was low-expressed in BC samples, and upregulation of miR-524-5p suppressed BC cell viability, migration, and invasion. FSTL1 was predicted as a target for miR-524-5p. In addition, overexpressed FSTL1 effectively abolished the effect of miR-524-5p on inhibiting FSTL1 expression, and reversed the inhibitory effects of miR-524-5p on the migration, invasion of BC cells as well as the effect of miR-524-5p on regulating the expressions of matrix metalloproteinase 2 (MMP2), matrix metalloproteinase 9 (MMP9), E-cadherin, and N-cadherin. Conclusions: Our findings suggest that miR-524-5p targeting FSTL1 adversely affects the progression of migration, invasion, and EMT of BC cells, thus, miR-524-5p is possibly a target for BC treatment.
Collapse
Affiliation(s)
- Taobo Jin
- Department of Thyroid and Breast Surgery, Zhuji People's Hospital, Zhuji City, China
| | - Yun Zhang
- Department of Thyroid and Breast Surgery, Zhuji People's Hospital, Zhuji City, China
| | - Tianya Zhang
- Department of Thyroid and Breast Surgery, Zhuji People's Hospital, Zhuji City, China
| |
Collapse
|
31
|
Kim WG, Cummings MC, Lakhani SR. Pitfalls and controversies in pathology impacting breast cancer management. Expert Rev Anticancer Ther 2020; 20:205-219. [PMID: 32174198 DOI: 10.1080/14737140.2020.1738222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Introduction: Breast cancer is a heterogeneous disease, at morphological, molecular, and clinical levels and this has significant implications for the diagnosis and management of the disease. The introduction of breast screening, and the use of small tissue sampling for diagnosis, the recognition of new morphological and molecular subtypes, and the increasing use of neoadjuvant therapies have created challenges in pathological diagnosis and classification.Areas covered: Areas of potential difficulty include columnar cell lesions, particularly flat epithelial atypia, atypical ductal hyperplasia, lobular neoplasia and its variants, and a range of papillary lesions. Fibroepithelial, sclerosing, mucinous, and apocrine lesions are also considered. Established and newer prognostic and predictive markers, such as immune infiltrates, PD-1 and PD-L1 and gene expression assays are evaluated. The unique challenges of pathology assessment post-neoadjuvant systemic therapy are also explored.Expert opinion: Controversies in clinical management arise due to incomplete and sometimes conflicting data on clinicopathological associations, prognosis, and outcome. The review will address some of these challenges.
Collapse
Affiliation(s)
- Woo Gyeong Kim
- Department of Pathology, University of Inje College of Medicine, Busan, Korea.,University of Queensland Centre for Clinical Research, Brisbane, Australia
| | - Margaret C Cummings
- University of Queensland Centre for Clinical Research, Brisbane, Australia.,Department of Anatomical Pathology, Pathology Queensland, Brisbane, Australia
| | - Sunil R Lakhani
- University of Queensland Centre for Clinical Research, Brisbane, Australia.,Department of Anatomical Pathology, Pathology Queensland, Brisbane, Australia
| |
Collapse
|
32
|
Al Amri WS, Allinson LM, Baxter DE, Bell SM, Hanby AM, Jones SJ, Shaaban AM, Stead LF, Verghese ET, Hughes TA. Genomic and Expression Analyses Define MUC17 and PCNX1 as Predictors of Chemotherapy Response in Breast Cancer. Mol Cancer Ther 2019; 19:945-955. [PMID: 31879365 DOI: 10.1158/1535-7163.mct-19-0940] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/12/2019] [Accepted: 12/17/2019] [Indexed: 12/24/2022]
Abstract
Poor-prognosis breast cancers are treated with cytotoxic chemotherapy, but often without any guidance from therapy predictive markers because universally accepted markers are not currently available. Treatment failure, in the form of recurrences, is relatively common. We aimed to identify chemotherapy predictive markers and resistance pathways in breast cancer. Our hypothesis was that tumor cells remaining after neoadjuvant chemotherapy (NAC) contain somatic variants causing therapy resistance, while variants present pre-NAC but lost post-NAC cause sensitivity. Whole-exome sequencing was performed on matched pre- and post-NAC cancer cells, which were isolated by laser microdissection, from 6 cancer cases, and somatic variants selected for or against by NAC were identified. Somatic variant diversity was significantly reduced after therapy (P < 0.05). MUC17 variants were identified in 3 tumors and were selected against by NAC in each case, while PCNX1 variants were identified in 2 tumors and were selected for in both cases, implicating the function of these genes in defining chemoresponse. In vitro knockdown of MUC17 or PCNX1 was associated with significantly increased or decreased chemotherapy sensitivity, respectively (P < 0.05), further supporting their roles in chemotherapy response. Expression was tested for predictive value in two independent cohorts of chemotherapy-treated breast cancers (n = 53, n = 303). Kaplan-Meier analyses revealed that low MUC17 expression was significantly associated with longer survival after chemotherapy, whereas low PCNX1 was significantly associated with reduced survival. We concluded that therapy-driven selection of somatic variants allows identification of chemotherapy response genes. With respect to MUC17 and PCNX1, therapy-driven selection acting on somatic variants, in vitro knockdown data concerning drug sensitivity, and survival analysis of expression levels in patient cohorts all define the genes as mediators of and predictive markers for chemotherapy response in breast cancer.
Collapse
Affiliation(s)
- Waleed S Al Amri
- School of Medicine, University of Leeds, Leeds, United Kingdom.,Department of Histopathology and Cytopathology, The Royal Hospital, Muscat, Oman
| | - Lisa M Allinson
- School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Diana E Baxter
- School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Sandra M Bell
- School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Andrew M Hanby
- School of Medicine, University of Leeds, Leeds, United Kingdom.,Department of Histopathology, St. James's University Hospital, Leeds, United Kingdom
| | - Stacey J Jones
- Department of Breast Surgery, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Abeer M Shaaban
- Histopathology and Cancer Sciences, Queen Elizabeth Hospital Birmingham and University of Birmingham, Birmingham, United Kingdom
| | - Lucy F Stead
- School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Eldo T Verghese
- Department of Histopathology, St. James's University Hospital, Leeds, United Kingdom
| | - Thomas A Hughes
- School of Medicine, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|