1
|
Chen KH, Barnes TA, Laskin J, Cheema P, Liu G, Iqbal M, Rothenstein J, Burkes R, Tsao MS, Leighl NB. The Perceived Value of Liquid Biopsy: Results From a Canadian Validation Study of Circulating Tumor DNA T790M Testing-Patient's Willingness-to-Pay: A Brief Report. JTO Clin Res Rep 2024; 5:100615. [PMID: 38292413 PMCID: PMC10826295 DOI: 10.1016/j.jtocrr.2023.100615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/06/2023] [Accepted: 11/27/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction Liquid biopsy is recommended to diagnose molecular resistance to targeted therapy in patients with lung cancer. Nevertheless, not all jurisdictions provide funding and patient access. We report patients' perceived value of liquid biopsy in targeted therapy resistance. Methods Canadian patients participating in a national EGFR T790M liquid biopsy validation study completed structured interviews measuring perceived value and willingness-to-pay for plasma circulating tumor DNA testing as an alternative to tumor biopsy using open-ended and iterative bidding approaches. Results A total of 60 patients with advanced lung cancer participated with a median age of 64 years (range: 31-87 y); 69% were Asian and 45% female. All had received prior EGFR tyrosine kinase inhibitor; 17% also received chemotherapy. All patients preferred to have plasma testing over repeat tumor biopsy. In the context of the Canadian publicly funded system, patients estimated that a median of 300 (interquartile range: 150-800) Canadian dollars was a reasonable price to pay for liquid biopsy. Patients were personally willing to pay a median 100 (interquartile range: 33-350) Canadian dollars. Conclusions In a system that covers the cost of standard diagnostic tests, patients with lung cancer indicated high willingness-to-pay out-of-pocket for liquid biopsy in the setting of acquired targeted therapy resistance. Patients have high perceived value of plasma genotyping and prefer it to repeat tumor biopsy.
Collapse
Affiliation(s)
- Kaitlin H. Chen
- Division of Medical Oncology/Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Tristan A. Barnes
- Medical Oncology, North Shore Private Hospital, St Leonard's, Australia
| | - Janessa Laskin
- Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Parneet Cheema
- Medical Oncology, William Osler Health System, Brampton, Ontario, Canada
| | - Geoffrey Liu
- Division of Medical Oncology/Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Mussawar Iqbal
- Medical Oncology, Allan Blair Cancer Centre, Regina, Saskatoon, Canada
| | | | - Ronald Burkes
- Medical Oncology, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Ming-Sound Tsao
- Division of Medical Oncology/Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
- Laboratory Medicine and Pathology, University Health Network, Toronto, Ontario, Canada
| | - Natasha B. Leighl
- Division of Medical Oncology/Hematology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Kramer A, Schuuring E, Vessies DCL, van der Leest P, Geerlings MJ, Rozendal P, Lanfermeijer M, Linders TC, van Kempen LC, Fijneman RJA, Ligtenberg MJL, Meijer GA, van den Broek D, Retèl VP, Coupé VMH. A Micro-Costing Framework for Circulating Tumor DNA Testing in Dutch Clinical Practice. J Mol Diagn 2023; 25:36-45. [PMID: 36402278 DOI: 10.1016/j.jmoldx.2022.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 07/11/2022] [Accepted: 10/04/2022] [Indexed: 11/18/2022] Open
Abstract
Circulating tumor DNA (ctDNA) is a promising new biomarker with multiple potential applications in cancer care. Estimating total cost of ctDNA testing is necessary for reimbursement and implementation, but challenging because of variations in workflow. We aimed to develop a micro-costing framework for consistent cost calculation of ctDNA testing. First, the foundation of the framework was built, based on the complete step-wise diagnostic workflow of ctDNA testing. Second, the costing method was set up, including costs for personnel, materials, equipment, overhead, and failures. Third, the framework was evaluated by experts and applied to six case studies, including PCR-, mass spectrometry-, and next-generation sequencing-based platforms, from three Dutch hospitals. The developed ctDNA micro-costing framework includes the diagnostic workflow from blood sample collection to diagnostic test result. The framework was developed from a Dutch perspective and takes testing volume into account. An open access tool is provided to allow for laboratory-specific calculations to explore the total costs of ctDNA testing specific workflow parameters matching the setting of interest. It also allows to straightforwardly assess the impact of alternative prices or assumptions on the cost per sample by simply varying the input parameters. The case studies showed a wide range of costs, from €168 to €7638 ($199 to $9124) per sample, and generated information. These costs are sensitive to the (coverage of) platform, setting, and testing volume.
Collapse
Affiliation(s)
- Astrid Kramer
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| | - Ed Schuuring
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Daan C L Vessies
- Department of Laboratory Medicine, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Paul van der Leest
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Pim Rozendal
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mirthe Lanfermeijer
- Department of Laboratory Medicine, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Theodora C Linders
- Department of Laboratory Medicine, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Léon C van Kempen
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Remond J A Fijneman
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marjolijn J L Ligtenberg
- Department of Human Genetics, Radboudumc, Nijmegen, the Netherlands; Department of Pathology, Radboudumc, Nijmegen, the Netherlands
| | - Gerrit A Meijer
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Daan van den Broek
- Department of Laboratory Medicine, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Valesca P Retèl
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Health Technology and Services Research, University of Twente, Enschede, the Netherlands
| | - Veerle M H Coupé
- Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| |
Collapse
|
3
|
García-Pardo M, Makarem M, Li JJN, Kelly D, Leighl NB. Integrating circulating-free DNA (cfDNA) analysis into clinical practice: opportunities and challenges. Br J Cancer 2022; 127:592-602. [PMID: 35347327 PMCID: PMC9381753 DOI: 10.1038/s41416-022-01776-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
In the current era of precision medicine, the identification of genomic alterations has revolutionised the management of patients with solid tumours. Recent advances in the detection and characterisation of circulating tumour DNA (ctDNA) have enabled the integration of liquid biopsy into clinical practice for molecular profiling. ctDNA has also emerged as a promising biomarker for prognostication, monitoring disease response, detection of minimal residual disease and early diagnosis. In this Review, we discuss current and future clinical applications of ctDNA primarily in non-small cell lung cancer in addition to other solid tumours.
Collapse
Affiliation(s)
- Miguel García-Pardo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Maisam Makarem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Janice J N Li
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Deirdre Kelly
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Natasha B Leighl
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
4
|
Makarem M, García-Pardo M, Leighl NB. Plasma-Based Genotyping in Advanced Solid Tumors: A Comprehensive Review. Cancers (Basel) 2021; 13:5299. [PMID: 34771462 PMCID: PMC8582457 DOI: 10.3390/cancers13215299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 12/25/2022] Open
Abstract
Molecular genotyping for advanced solid malignancies has transformed the clinical management of patients with metastatic disease. Treatment decisions in a growing number of tumors require knowledge of molecularly driven alterations in order to select optimal targeted therapy. Although genomic testing of tumor tissue is the gold standard for identifying targetable genomic alterations, biopsy samples are often limited or difficult to access. This has paved the way for the development of plasma-based approaches for genomic profiling. Recent advances in the detection of plasma-circulating tumor DNA (ctDNA) have enabled the integration of plasma-based molecular profiling into clinical practice as an alternative or complementary tool for genomic testing in the setting of advanced cancer, to facilitate the identification of driver mutations to guide initial treatment and diagnose resistance. Several guidelines now recommend the use of plasma where tumor tissue is limited to identify a targetable genomic alteration. Current plasma-based assays can evaluate multiple genes in comprehensive panels, and their application in advanced disease will be increasingly incorporated into standard practice. This review focuses on current and future applications of plasma ctDNA-based assays in advanced solid malignancies, while highlighting some limitations in implementing this technology into clinical practice.
Collapse
Affiliation(s)
| | | | - Natasha B. Leighl
- Princess Margaret Cancer Center, Department of Medical Oncology, Toronto, ON M5G 2C1, Canada; (M.M.); (M.G.-P.)
| |
Collapse
|
5
|
Makarem M, Ezeife DA, Smith AC, Li JJN, Law JH, Tsao MS, Leighl NB. Reflex ROS1 IHC Screening with FISH Confirmation for Advanced Non-Small Cell Lung Cancer-A Cost-Efficient Strategy in a Public Healthcare System. Curr Oncol 2021; 28:3268-3279. [PMID: 34449580 PMCID: PMC8395515 DOI: 10.3390/curroncol28050284] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/11/2022] Open
Abstract
ROS1 rearrangements are identified in 1-2% of lung adenocarcinoma cases, and reflex testing is guideline-recommended. We developed a decision model for population-based ROS1 testing from a Canadian public healthcare perspective to determine the strategy that optimized detection of true-positive (TP) cases while minimizing costs and turnaround time (TAT). Eight diagnostic strategies were compared, including reflex single gene testing via immunohistochemistry (IHC) screening, fluorescence in-situ hybridization (FISH), next-generation sequencing (NGS), and biomarker-informed (EGFR/ALK/KRAS wildtype) testing initiated by pathologists and clinician-initiated strategies. Reflex IHC screening with FISH confirmation of positive cases yielded the best results for TAT, TP detection rate, and cost. IHC screening saved CAD 1,000,000 versus reflex FISH testing. NGS was the costliest reflex strategy. Biomarker-informed testing was cost-efficient but delayed TAT. Clinician-initiated testing was the least costly but resulted in long TAT and missed TP cases, highlighting the importance of reflex testing. Thus, reflex IHC screening for ROS1 with FISH confirmation provides a cost-efficient strategy with short TAT and maximizes the number of TP cases detected.
Collapse
Affiliation(s)
- Maisam Makarem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; (M.M.); (A.C.S.); (J.J.N.L.); (J.H.L.); (M.-S.T.)
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Doreen A. Ezeife
- Tom Baker Cancer Centre, University of Calgary, Calgary, AB T2N 4N2, Canada;
| | - Adam C. Smith
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; (M.M.); (A.C.S.); (J.J.N.L.); (J.H.L.); (M.-S.T.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Janice J. N. Li
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; (M.M.); (A.C.S.); (J.J.N.L.); (J.H.L.); (M.-S.T.)
| | - Jennifer H. Law
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; (M.M.); (A.C.S.); (J.J.N.L.); (J.H.L.); (M.-S.T.)
| | - Ming-Sound Tsao
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; (M.M.); (A.C.S.); (J.J.N.L.); (J.H.L.); (M.-S.T.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Natasha B. Leighl
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; (M.M.); (A.C.S.); (J.J.N.L.); (J.H.L.); (M.-S.T.)
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
6
|
Selvarajah S, Plante S, Speevak M, Vaags A, Hamelinck D, Butcher M, McCready E, Grafodatskaya D, Blais N, Tran-Thanh D, Weng X, Nassabein R, Greer W, Walton RN, Lo B, Demetrick D, Santos S, Sadikovic B, Zhang X, Zhang T, Spence T, Stockley T, Feilotter H, Joubert P. A Pan-Canadian Validation Study for the Detection of EGFR T790M Mutation Using Circulating Tumor DNA From Peripheral Blood. JTO Clin Res Rep 2021; 2:100212. [PMID: 34590051 PMCID: PMC8474449 DOI: 10.1016/j.jtocrr.2021.100212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/23/2021] [Accepted: 07/08/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Genotyping circulating tumor DNA (ctDNA) is a promising noninvasive clinical tool to identify the EGFR T790M resistance mutation in patients with advanced NSCLC with resistance to EGFR inhibitors. To facilitate standardization and clinical adoption of ctDNA testing across Canada, we developed a 2-phase multicenter study to standardize T790M mutation detection using plasma ctDNA testing. METHODS In phase 1, commercial reference standards were distributed to participating clinical laboratories, to use their existing platforms for mutation detection. Baseline performance characteristics were established using known and blinded engineered plasma samples spiked with predetermined concentrations of T790M, L858R, and exon 19 deletion variants. In phase II, peripheral blood collected from local patients with known EGFR activating mutations and progressing on treatment were assayed for the presence of EGFR variants and concordance with a clinically validated test at the reference laboratory. RESULTS All laboratories in phase 1 detected the variants at 0.5 % and 5.0 % allele frequencies, with no false positives. In phase 2, the concordance with the reference laboratory for detection of both the primary and resistance mutation was high, with next-generation sequencing and droplet digital polymerase chain reaction exhibiting the best overall concordance. Data also suggested that the ability to detect mutations at clinically relevant limits of detection is generally not platform-specific, but rather impacted by laboratory-specific practices. CONCLUSIONS Discrepancies among sending laboratories using the same assay suggest that laboratory-specific practices may impact performance. In addition, a negative or inconclusive ctDNA test should be followed by tumor testing when possible.
Collapse
Affiliation(s)
- Shamini Selvarajah
- Department of Laboratory Medicine and Genetics, Trillium Health Partners, Mississauga, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Sophie Plante
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Québec, Quebec, Canada
| | - Marsha Speevak
- Department of Laboratory Medicine and Genetics, Trillium Health Partners, Mississauga, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Andrea Vaags
- Department of Laboratory Medicine and Genetics, Trillium Health Partners, Mississauga, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Darren Hamelinck
- Department of Laboratory Medicine and Genetics, Trillium Health Partners, Mississauga, Ontario, Canada
| | - Martin Butcher
- Department of Oncology, McMaster University, Hamilton, Ontario, Canada
| | - Elizabeth McCready
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Hamilton Regional Laboratory Medicine Program, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Daria Grafodatskaya
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
- Hamilton Regional Laboratory Medicine Program, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Normand Blais
- Centre Hospitalier de l’Université de Montréal, Montréal, Quebec, Canada
| | - Danh Tran-Thanh
- Centre Hospitalier de l’Université de Montréal, Montréal, Quebec, Canada
| | - Xiaoduan Weng
- Centre Hospitalier de l’Université de Montréal, Montréal, Quebec, Canada
| | - Rami Nassabein
- Centre Hospitalier de l’Université de Montréal, Montréal, Quebec, Canada
| | - Wenda Greer
- Queen Elizabeth II Health Sciences Center, Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | - Bryan Lo
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Doug Demetrick
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Oncology, University of Calgary, Calgary, Alberta, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- Department of Medical Genetics, University of Calgary, Calgary, Alberta, Canada
| | - Stephanie Santos
- Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences Centre, London, Ontario, Canada
| | - Bekim Sadikovic
- Molecular Genetics Laboratory, Molecular Diagnostics Division, London Health Sciences Centre, London, Ontario, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, Ontario, Canada
| | - Xiao Zhang
- Laboratory Genetics, Kingston Health Sciences Center, Kingston, Ontario, Canada
| | - Tong Zhang
- Department of Clinical Laboratory Genetics, University Health Network, Toronto, Ontario, Canada
| | - Tara Spence
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, Ontario, Canada
| | - Tracy Stockley
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Clinical Laboratory Genetics, University Health Network, Toronto, Ontario, Canada
| | - Harriet Feilotter
- Laboratory Genetics, Kingston Health Sciences Center, Kingston, Ontario, Canada
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, Ontario, Canada
| | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Québec-Université Laval, Québec, Quebec, Canada
| |
Collapse
|
7
|
Park C, Lee S, Lee JC, Choi C, Lee SY, Jang T, Oh I, Kim Y. Phase II open-label multicenter study to assess the antitumor activity of afatinib in lung cancer patients with activating epidermal growth factor receptor mutation from circulating tumor DNA: Liquid-Lung-A. Thorac Cancer 2021; 12:444-452. [PMID: 33270375 PMCID: PMC7882376 DOI: 10.1111/1759-7714.13763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/10/2020] [Accepted: 11/15/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Mutation analysis of circulating tumor DNA (ctDNA) is used for diagnosing lung cancer. This trial aimed to assess the efficacy of afatinib in treatment-naïve patients with lung cancer harboring epidermal growth factor receptor mutations (EGFRm, exon 19 deletions or exon 21 point mutations) detected based on ctDNA. METHODS The primary objective was the objective response rate (ORR) in the response evaluable (RE) population. EGFRm analysis of ctDNA was performed using PANA Mutype. Of the 331 patients screened, ctDNA was positive in 21% (68/331) in the detection of activating EGFRm. Among 81 subjects with tumor EGFRm, 48 showed matched EGFRm in their ctDNA (59% sensitivity). RESULTS Therapy with afatinib 40 mg was initiated in 21 (female, 17; adenocarcinoma, 20) patients (intention-to-treat, ITT); dose modifications were made in 15 (71%). The ORR was 74% in the RE population (14/19); 11 patients showed EGFRm only in ctDNA (Group A), whereas 10 exhibited the same EGFRm in their ctDNA and tumor DNA (Group B). There was no significant difference in ORR between Groups A and B (80% and 67% RE, respectively). Median progression-free survival (PFS) was 12.0 months, and no significant difference was observed according to the final afatinib dose, type of EGFRm, and Group A versus B. After progression, T790M mutation was found in 40% (6/15) of patients, and osimertinib was used as a second-line treatment. CONCLUSIONS Afatinib showed similar ORR and PFS in patients with lung cancer harboring EGFRm in their ctDNA regardless of tumor EGFRm results. KEY POINTS SIGNIFICANT FINDINGS OF THE STUDY: Afatinib showed favorable ORR and PFS regardless of the tumor EGFR mutation status results, similar to the findings of previous trials assessing afatinib as first-line treatment of EGFR-mutated non-small cell lung cancer based on tumor genotyping. WHAT THIS STUDY ADDS Our findings emphasize that the survival benefit of afatinib treatment can be achieved not only by appropriate dose reduction with frequent and detailed monitoring of toxicities, but also by using noninvasive (ctDNA) assays in a real-world setting.
Collapse
Affiliation(s)
- Cheol‐Kyu Park
- Department of Internal MedicineChonnam National University Medical School and CNU Hwasun HospitalHwasunJeonnamKorea
| | - Sung‐Yong Lee
- Department of Internal MedicineKorea University Guro HospitalSeoulKorea
| | - Jae Cheol Lee
- Department of Oncology, Pulmonary and Critical Care MedicineCollege of Medicine, University of Ulsan, Asan Medical CenterSeoulKorea
| | - Chang‐Min Choi
- Department of Oncology, Pulmonary and Critical Care MedicineCollege of Medicine, University of Ulsan, Asan Medical CenterSeoulKorea
| | - Shin Yup Lee
- Department of Internal Medicine, School of MedicineKyungpook National UniversityDaeguKorea
| | - Tae‐Won Jang
- Department of Internal Medicine, School of MedicineKosin University Gospel HospitalPusanKorea
| | - In‐Jae Oh
- Department of Internal MedicineChonnam National University Medical School and CNU Hwasun HospitalHwasunJeonnamKorea
| | - Young‐Chul Kim
- Department of Internal MedicineChonnam National University Medical School and CNU Hwasun HospitalHwasunJeonnamKorea
| |
Collapse
|