101
|
Tan AC, Tan DSW. Targeted Therapies for Lung Cancer Patients With Oncogenic Driver Molecular Alterations. J Clin Oncol 2022; 40:611-625. [PMID: 34985916 DOI: 10.1200/jco.21.01626] [Citation(s) in RCA: 284] [Impact Index Per Article: 142.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Lung cancer has traditionally been classified by histology. However, a greater understanding of disease biology and the identification of oncogenic driver alterations has dramatically altered the therapeutic landscape. Consequently, the new classification paradigm of non-small-cell lung cancer is further characterized by molecularly defined subsets actionable with targeted therapies and the treatment landscape is becoming increasingly complex. This review encompasses the current standards of care for targeted therapies in lung cancer with driver molecular alterations. Targeted therapies for EGFR exon 19 deletion and L858R mutations, and ALK and ROS1 rearrangements are well established. However, there is an expanding list of approved targeted therapies including for BRAF V600E, EGFR exon 20 insertion, and KRAS G12C mutations, MET exon 14 alterations, and NTRK and RET rearrangements. In addition, there are numerous other oncogenic drivers, such as HER2 exon 20 insertion mutations, for which there are emerging efficacy data for targeted therapies. The importance of diagnostic molecular testing, intracranial efficacy of novel therapies, the optimal sequencing of therapies, role for targeted therapies in early-stage disease, and future directions for precision oncology approaches to understand tumor evolution and therapeutic resistance are also discussed.
Collapse
Affiliation(s)
- Aaron C Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore.,Duke-NUS Medical School, National University of Singapore, Singapore
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore.,Duke-NUS Medical School, National University of Singapore, Singapore.,Genome Institute of Singapore, Singapore
| |
Collapse
|
102
|
Perdrizet K, Stockley TL, Law JH, Smith A, Zhang T, Fernandes R, Shabir M, Sabatini P, Youssef NA, Ishu C, Li JJ, Tsao MS, Pal P, Cabanero M, Schwock J, Ko HM, Boerner S, Ruff H, Shepherd FA, Bradbury PA, Liu G, Sacher AG, Leighl NB. Integrating comprehensive genomic sequencing of non-small cell lung cancer into a public healthcare system. Cancer Treat Res Commun 2022; 31:100534. [PMID: 35278845 DOI: 10.1016/j.ctarc.2022.100534] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 06/14/2023]
Abstract
OBJECTIVES Standard molecular testing for patients with stage IV non-small cell lung cancer (NSCLC) in the Canadian publicly funded health system includes single gene testing for EGFR, ALK, and ROS-1. Comprehensive genomic profiling (CGP) may broaden treatment options for patients. This study examined the impact of CGP in a publicly funded health system. METHODS Consenting patients with stage IV NSCLC without known targetable alterations underwent CGP on diagnostic samples. Patients that had progressed on targeted therapy were also eligible. The CGP assay was a hybrid capture next generation sequencing (NGS) panel (Oncomine Comprehensive Assay Version 3, ThermoFisher). The number of actionable alterations, changes in treatment, clinical trial eligibility and costs as a result of CGP were evaluated and patient willingness-to-pay. RESULTS Of 182 screened patients,134 (74%) had successful CGP testing. Twenty percent had received prior targeted therapy. Incremental actionable alterations were identified in 31% of patients. The most common novel targets identified were mutations in ERBB2 (exon 20 insertions), MET (exon 14 skipping) and KRAS (G12C). At data cut off (31/12/2020), 16% of patients had a change in treatment as a result of CGP. Additional clinical trial options were identified for 75% of patients. The incremental direct laboratory cost for CGP beyond public reimbursement for single gene tests was $747 CAD/case. CONCLUSION CGP identifies additional actionable targets beyond single gene tests with a direct impact on patient treatment and increased clinical trial eligibility. These benefits highlight the value of CGP in patients with NSCLC in public health systems.
Collapse
Affiliation(s)
- Kirstin Perdrizet
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada; William Osler Health System, Brampton, Ontario, Canada.
| | - Tracy L Stockley
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada; Advanced Molecular Diagnostics Laboratory, University Health Network, Toronto, Canada
| | - Jennifer H Law
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada
| | - Adam Smith
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada; Advanced Molecular Diagnostics Laboratory, University Health Network, Toronto, Canada
| | - Tong Zhang
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada; Advanced Molecular Diagnostics Laboratory, University Health Network, Toronto, Canada
| | - Roxanne Fernandes
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada
| | - Muqdas Shabir
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada
| | - Peter Sabatini
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada; Advanced Molecular Diagnostics Laboratory, University Health Network, Toronto, Canada
| | - Nadia Al Youssef
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada; Advanced Molecular Diagnostics Laboratory, University Health Network, Toronto, Canada
| | - Christine Ishu
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada; Advanced Molecular Diagnostics Laboratory, University Health Network, Toronto, Canada
| | - Janice Jn Li
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada
| | - Ming-Sound Tsao
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada
| | - Prodipto Pal
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada
| | - Michael Cabanero
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada
| | - Joerg Schwock
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada
| | - Hyang Mi Ko
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada
| | - Scott Boerner
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada
| | - Heather Ruff
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada
| | - Frances A Shepherd
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada
| | | | - Geoffrey Liu
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada
| | - Adrian G Sacher
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada
| | - Natasha B Leighl
- Princess Margaret Cancer Centre/University Health Network, Toronto, Canada.
| |
Collapse
|
103
|
Corral de la Fuente E, Olmedo Garcia ME, Gomez Rueda A, Lage Y, Garrido P. Targeting KRAS in Non-Small Cell Lung Cancer. Front Oncol 2022; 11:792635. [PMID: 35083149 PMCID: PMC8784727 DOI: 10.3389/fonc.2021.792635] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Kirsten Rat Sarcoma viral oncogene homolog (KRAS) is the most frequently altered oncogene in Non-Small Cell Lung Cancer (NSCLC). KRAS mutant tumors constitute a heterogeneous group of diseases, different from other oncogene-derived tumors in terms of biology and response to treatment, which hinders the development of effective drugs against KRAS. Therefore, for decades, despite enormous efforts invested in the development of drugs aimed at inhibiting KRAS or its signaling pathways, KRAS was considered to be undruggable. Recently, the discovery of a new pocket under the effector binding switch II region of KRAS G12C has allowed the development of direct KRAS inhibitors such as sotorasib, the first FDA-approved drug targeting KRAS G12C, or adagrasib, initiating a new exciting era. However, treatment with targeted KRAS G12C inhibitors also leads to resistance, and understanding the possible mechanisms of resistance and which drugs could be useful to overcome it is key. Among others, KRAS G12C (ON) tricomplex inhibitors and different combination therapy strategies are being analyzed in clinical trials. Another area of interest is the potential role of co-mutations in treatment selection, particularly immunotherapy. The best first-line strategy remains to be determined and, due to the heterogeneity of KRAS, is likely to be based on combination therapies.
Collapse
Affiliation(s)
- Elena Corral de la Fuente
- Early Phase Clinical Drug Development in Oncology, South Texas Accelerated Research Therapeutics (START) Madrid-Centro Integral Oncológico Clara Campal (CIOCC), Centro Integral Oncológico Clara Campal, Madrid, Spain
| | | | - Ana Gomez Rueda
- Department of Medical Oncology, Ramón y Cajal University Hospital, Madrid, Spain
| | - Yolanda Lage
- Department of Medical Oncology, Ramón y Cajal University Hospital, Madrid, Spain
| | - Pilar Garrido
- Department of Medical Oncology, Ramón y Cajal University Hospital, Madrid, Spain
| |
Collapse
|
104
|
Chen S, Fu A, Lu Y, Lu W, Chen Y, Hong S, Zhou S, Xiang T, Zhang Z, Cai Y. Investigating the genomic alteration improved the clinical outcome of aged patients with lung carcinoma. BMC Genomics 2022; 23:55. [PMID: 35031014 PMCID: PMC8760649 DOI: 10.1186/s12864-021-08289-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/28/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lung carcinoma is a common geriatric disease. The development of genotype-targeted therapies greatly improved the management of lung carcinoma. However, the treatment for old patients can be more complex than that for young individuals. RESULTS To investigate the benefits of genetic detection for older patients with lung carcinoma, we explored the genomic profiling of 258 patients with more than 55 years using a targeted next generation sequencing, and some of these patients were treated with targeted therapies based on the results of genomic detection. KRAS codon 61 mutations were found in 15.2% KRAS-mutated patients, which tend to be co-existing with other classical activating mutations other than codons 12/13. Acquired EGFR C797S mutations were identified in 2 cases and ERBB2 amplification was identified in 1 case. All these 3 cases developed resistance to EGFR tyrosine kinase inhibitors and showed expected results of their followed therapies. The median progression-free survival and median overall survival of patients treated with molecular targeted therapies were better than those of patients treated with chemoradiotherapy alone. CONCLUSIONS Our findings revealed the specific genomic profiles of patients older than 55 years with lung carcinoma and suggested that these old patients have been benefit from the genetic detection, which helped identify druggable mutations and distinguish resistance mechanisms.
Collapse
Affiliation(s)
- Sixian Chen
- Medical Oncology Department V, Guangdong Nongken Central Hospital, Zhanjiang, 524002, China
| | - Aizhen Fu
- Gynecology Department, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yuan Lu
- Medical Oncology Department V, Guangdong Nongken Central Hospital, Zhanjiang, 524002, China
| | - Wei Lu
- Medical Oncology Department V, Guangdong Nongken Central Hospital, Zhanjiang, 524002, China
| | - Yongfeng Chen
- Medical Oncology Department V, Guangdong Nongken Central Hospital, Zhanjiang, 524002, China
| | - Shuiqiang Hong
- Medical Oncology Department V, Guangdong Nongken Central Hospital, Zhanjiang, 524002, China
| | - Suli Zhou
- Medical Oncology Department V, Guangdong Nongken Central Hospital, Zhanjiang, 524002, China
| | | | | | - Yongguang Cai
- Medical Oncology Department V, Guangdong Nongken Central Hospital, Zhanjiang, 524002, China.
| |
Collapse
|
105
|
Bourhis A, Remoué A, Samaison L, Uguen A. Diagnostic mutationnel rapide Idylla™ : applications théranostiques actuelles et futures. Ann Pathol 2022; 42:329-343. [DOI: 10.1016/j.annpat.2021.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 12/07/2021] [Accepted: 12/12/2021] [Indexed: 10/19/2022]
|
106
|
Zhao JJ, Ping Chan H, Yang Soon Y, Huang Y, Soo RA, Kee AC. A systematic review and meta-analysis of the adequacy of endobronchial ultrasound transbronchial needle aspiration for next-generation sequencing in patients with non-small cell lung cancer. Lung Cancer 2022; 166:17-26. [DOI: 10.1016/j.lungcan.2022.01.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 01/14/2022] [Accepted: 01/20/2022] [Indexed: 12/17/2022]
|
107
|
Direct identification of ALK and ROS1 fusions in non-small cell lung cancer from hematoxylin and eosin-stained slides using deep learning algorithms. Mod Pathol 2022; 35:1882-1887. [PMID: 36057739 PMCID: PMC9708557 DOI: 10.1038/s41379-022-01141-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022]
Abstract
Anaplastic lymphoma kinase (ALK) and ROS oncogene 1 (ROS1) gene fusions are well-established key players in non-small cell lung cancer (NSCLC). Although their frequency is relatively low, their detection is important for patient care and guides therapeutic decisions. The accepted methods used for their detection are immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) assay, as well as DNA and RNA-based sequencing methodologies. These assays are expensive, time-consuming, and require technical expertise and specialized equipment as well as biological specimens that are not always available. Here we present an alternative detection method using a computer vision deep learning approach. An advanced convolutional neural network (CNN) was used to generate classifier models to detect ALK and ROS1-fusions directly from scanned hematoxylin and eosin (H&E) whole slide images prepared from NSCLC tumors of patients. A two-step training approach was applied, with an initial unsupervised training step performed on a pan-cancer sample cohort followed by a semi-supervised fine-tuning step, which supported the development of a classifier with performances equal to those accepted for diagnostic tests. Validation of the ALK/ROS1 classifier on a cohort of 72 lung cancer cases who underwent ALK and ROS1-fusion testing at the pathology department at Sheba Medical Center displayed sensitivities of 100% for both genes (six ALK-positive and two ROS1-positive cases) and specificities of 100% and 98.6% respectively for ALK and ROS1, with only one false-positive result for ROS1-alteration. These results demonstrate the potential advantages that machine learning solutions may have in the molecular pathology domain, by allowing fast, standardized, accurate, and robust biomarker detection overcoming many limitations encountered when using current techniques. The integration of such novel solutions into the routine pathology workflow can support and improve the current clinical pipeline.
Collapse
|
108
|
Martín-López J, Rojo F, Martínez-Pozo A, Hernández-Iglesias T, Carcedo D, de Alda LR, García JF, Salas C. Biomarker testing strategies in non-small cell lung cancer in the real-world setting: analysis of methods in the Prospective Central Lung Cancer Biomarker Registry (LungPath) from the Spanish Society of Pathology (SEAP). J Clin Pathol 2021; 76:327-332. [PMID: 34903610 PMCID: PMC10176377 DOI: 10.1136/jclinpath-2021-208034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/09/2021] [Indexed: 11/04/2022]
Abstract
AIMS The aim of this study is to extend the analysis of the Lung Cancer Biomarker Testing Registry (LungPath), by analysing the techniques used in the determination of epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), c-ros oncogene 1 (ROS1) and programmed death ligand-1 (PD-L1) for the diagnostic of patients with advanced non-small-cell lung cancer (NSCLC). METHODS Information of the technique used for the determination of EGFR, ALK, ROS1 and PD-L1 was recorded from March 2018 to January 2019 from 44 centres, but only 34 centres matched with the 38 centres previously analysed, allowing to analyse the techniques used in 8970 matched determinations of EGFR, ALK, ROS1 and PD-L1. Therefore, a by-centre analysis studied the level of implementation of the techniques in the 44 centres, while a by-determination analysis made it possible to assess the overall frequency of the techniques used on the 9134 matched samples. RESULTS By-centre analysis showed that only 46.5% and 25.6% of the centres used reflex strategies for ALK and ROS1 determination, respectively. By-determination analysis showed that 94.4% of EGFR determinations were performed by PCR, 80.7% of ALK determinations were performed by IHC with clone D5F3, while 55.7% of ROS1 determinations were performed by IHC with clone D4D6. 22C3 were the PD-L1 clone more used (43.5%) followed by SP263 clone (31.1%). CONCLUSIONS The real-world evidence obtained from LungPath shows the effort of Spanish hospitals in performing biomarker determination in NSCLC with different methodologies despite that next-generation sequencing (NGS) utilisation in the year of the analysis was low. Biomarker determination results could be optimised with the incorporation of sequencing methods such as NGS in pathology departments.
Collapse
Affiliation(s)
- Javier Martín-López
- Pathology Department, Hospital Universitario Puerta del Hierro Majadahonda, Madrid, Spain
| | - Federico Rojo
- Pathology Department, Hospital Universitario Fundacion Jimenez Diaz-CIBERONC, Madrid, Spain.,External Quality Asessment (GCP), Spanish Society of Anatomical Pathology, Madrid, Spain
| | - Antonio Martínez-Pozo
- External Quality Asessment (GCP), Spanish Society of Anatomical Pathology, Madrid, Spain.,Pathology Department, Hospital Clinic de Barcelona, Barcelona, Spain
| | | | | | | | | | - Clara Salas
- Pathology Department, Hospital Universitario Puerta del Hierro Majadahonda, Madrid, Spain.,External Quality Asessment (GCP), Spanish Society of Anatomical Pathology, Madrid, Spain
| |
Collapse
|
109
|
Zheng Y, Vioix H, Liu FX, Singh B, Sharma S, Sharda D. Diagnostic and economic value of biomarker testing for targetable mutations in non-small-cell lung cancer: a literature review. Future Oncol 2021; 18:505-518. [PMID: 34865516 DOI: 10.2217/fon-2021-1040] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We aimed to assess the diagnostic and economic value of next-generation sequencing (NGS) versus single-gene testing, and of liquid biopsy (LBx) versus tissue biopsy (TBx) in non-small-cell lung cancer biomarker testing through literature review. Embase and MEDLINE were searched to identify relevant studies (n = 43) from 2015 to 2020 in adults with advanced non-small-cell lung cancer. For NGS versus single-gene testing, concordance was 70-99% and sensitivity was 86-100%. For LBx versus TBx, specificity was 43-100% and sensitivity was ≥60%. Turnaround times were longer for NGS versus single-gene testing (but not vs sequential testing) and faster for LBx versus TBx. NGS was cost-effective, and LBx reduced US per-patient costs. NGS versus single-gene testing and LBx versus TBx were concordant. NGS and LBx may be cost-effective for initial screening.
Collapse
Affiliation(s)
- Ying Zheng
- US Health Economics and Outcomes Research, EMD Serono, Inc., An affiliate of Merck KGaA, Rockland, MA 02370, USA
| | - Helene Vioix
- Global Evidence & Value Development, Merck Healthcare KGaA, Darmstadt, Germany
| | - Frank X Liu
- US Health Economics and Outcomes Research, EMD Serono, Inc., An affiliate of Merck KGaA, Rockland, MA 02370, USA
| | | | - Sakshi Sharma
- HEOR, Parexel, Access Consulting, Mohali, Punjab, India
| | - Deepti Sharda
- HEOR, Parexel, Access Consulting, Mohali, Punjab, India
| |
Collapse
|
110
|
Cheng Y, Zhang T, Xu Q. Therapeutic advances in non-small cell lung cancer: Focus on clinical development of targeted therapy and immunotherapy. MedComm (Beijing) 2021; 2:692-729. [PMID: 34977873 PMCID: PMC8706764 DOI: 10.1002/mco2.105] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 02/05/2023] Open
Abstract
Lung cancer still contributes to nearly one-quarter cancer-related deaths in the past decades, despite the rapid development of targeted therapy and immunotherapy in non-small cell lung cancer (NSCLC). The development and availability of comprehensive genomic profiling make the classification of NSCLC more precise and personalized. Most treatment decisions of advanced-stage NSCLC have been made based on the genetic features and PD-L1 expression of patients. For the past 2 years, more than 10 therapeutic strategies have been approved as first-line treatment for certain subgroups of NSCLC. However, some major challenges remain, including drug resistance and low rate of overall survival. Therefore, we discuss and review the therapeutic strategies of NSCLC, and focus on the development of targeted therapy and immunotherapy in advanced-stage NSCLC. Based on the latest guidelines, we provide an updated summary on the standard treatment for NSCLC. At last, we discussed several potential therapies for NSCLC. The development of new drugs and combination therapies both provide promising therapeutic effects on NSCLC.
Collapse
Affiliation(s)
- Yuan Cheng
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Tao Zhang
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of Biotherapy and Cancer CenterNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Qing Xu
- Department of OncologyShanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| |
Collapse
|
111
|
Fox AH, Jett JR, Roy UB, Johnson BE, King JC, Martin N, Osarogiagbon RU, Rivera MP, Rosenthal LS, Smith RA, Silvestri GA. Knowledge and Practice Patterns Among Pulmonologists for Molecular Biomarker Testing in Advanced Non-small Cell Lung Cancer. Chest 2021; 160:2293-2303. [PMID: 34181954 PMCID: PMC8727850 DOI: 10.1016/j.chest.2021.06.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Targeted therapies for advanced non-small cell lung cancer (NSCLC) with oncogenic drivers have caused a paradigm shift in care. Biomarker testing is needed to assess eligibility for these therapies. Pulmonologists often perform bronchoscopy, providing tissue for both pathologic diagnosis and biomarker analysis. We performed this survey to define the existing knowledge and practices regarding the pulmonologists' role in biomarker testing for advanced NSCLC. RESEARCH QUESTION What is the current knowledge and practice of pulmonologists regarding biomarker testing and targeted therapies in advanced NSCLC? STUDY DESIGN AND METHODS This cross-sectional study was performed using an electronic survey of a random sample of 7,238 pulmonologists. Questions focused on diagnostic steps and biomarker analyses for NSCLC. RESULTS A total of 453 pulmonologists responded. Respondents vary by reported lung cancer patient volume, ranging from 51% evaluating one to four new cases per month to 19% evaluating > 10 cases per month. Interventional training, academic practice setting, and higher volume of endobronchial ultrasound with transbronchial needle aspiration (EBUS-TBNA) were associated with increased knowledge of practice guidelines for the number of recommended passes during EBUS-TBNA (P < .05). Academic pulmonologists more commonly performed or referred for EBUS-TBNA than community pulmonologists (96% and 83%, respectively; P < .0005). Higher testing rates were associated with interventional training, academic setting, and the presence of an institutional policy, whereas lower testing rates were associated with general pulmonologists, practice in community settings, and lack of a guiding institutional policy (P < .05). INTERPRETATION Substantial differences among pulmonologists' evaluation of advanced NSCLC, variation in knowledge of available biomarkers and the importance of targeted therapies, and differences in institutional coordination likely lead to underutilization of biomarker testing. Interventional training appears to drive improved knowledge and practice for biomarker testing more than practice setting. Improvements are needed in tissue acquisition and interdisciplinary coordination to ensure universal and comprehensive testing for eligible patients.
Collapse
Affiliation(s)
- Adam H Fox
- Division of Pulmonary and Critical Care Medicine, Medical University of South Carolina, Charleston, SC
| | | | | | | | | | | | | | - M Patricia Rivera
- Division of Pulmonary and Critical Care Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Lauren S Rosenthal
- Prevention and Early Detection Department, American Cancer Society, Atlanta, GA
| | - Robert A Smith
- Prevention and Early Detection Department, American Cancer Society, Atlanta, GA
| | - Gerard A Silvestri
- Division of Pulmonary and Critical Care Medicine, Medical University of South Carolina, Charleston, SC.
| |
Collapse
|
112
|
Buglioni S, Marino M. Pitfalls in Molecular Testing and the Added Value of the Cancer Research Biomedical Community. J Thorac Oncol 2021; 16:1976-1978. [PMID: 34809799 DOI: 10.1016/j.jtho.2021.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 09/09/2021] [Indexed: 01/20/2023]
Affiliation(s)
- Simonetta Buglioni
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Mirella Marino
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
113
|
Pelosi G. KEAP1 and TP53 (Co)mutation in Lung Adenocarcinoma: Another Bullet for Immunotherapy? J Thorac Oncol 2021; 16:1979-1983. [PMID: 34809800 DOI: 10.1016/j.jtho.2021.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 01/21/2023]
Affiliation(s)
- Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Inter-Hospital Pathology Division, Istituto di Ricovero e Cura a Carattere Scientifico MultiMedica, Milan, Italy.
| |
Collapse
|
114
|
Worldwide Prevalence of Epidermal Growth Factor Receptor Mutations in Non-Small Cell Lung Cancer: A Meta-Analysis. Mol Diagn Ther 2021; 26:7-18. [PMID: 34813053 PMCID: PMC8766385 DOI: 10.1007/s40291-021-00563-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2021] [Indexed: 01/22/2023]
Abstract
Background Identification of variable epidermal growth factor receptor (EGFR) gene mutations in non-small cell lung cancer (NSCLC) is important for the selection of appropriate targeted therapies. This meta-analysis was conducted to provide a worldwide overview of EGFR mutation and submutation (specifically exon 19 deletions, exon 21 L858R substitutions, and others) prevalence, and identify important covariates that influence EGFR mutation status in patients with advanced NSCLC to address this clinical data gap. Methods Embase® and MEDLINE® in Ovid were searched for studies published between 2004 and 2019 with cohorts of ≥ 50 adults with EGFR mutations, focusing on stage III/IV NSCLC (≤ 20% of patients with stage I/II NSCLC). Linear mixed-effects models were fitted to EGFR mutation endpoints using logistic transformation (logit), assuming a binomial distribution. The model included terms for an intercept reflecting European studies and further additive terms for other continents. EGFR submutations examined were exon 19 deletions, exon 21 L858R substitutions, and others. Results Of 3969 abstracts screened, 57 studies were included in the overall EGFR mutation analysis and 74 were included in the submutation analysis relative to the overall EGFR mutation population (Europe, n = 12; Asia, n = 51; North America, n = 5; Central America, n = 1; South America, n = 1; Oceania, n = 1; Global, n = 3). The final overall EGFR mutations model estimated Asian and European prevalence of 49.1% and 12.8%, respectively, and included an additive covariate for the proportion of male patients in a study. There were no significant covariates in the submutation analyses. Most submutations were actionable: exon 19 deletions (49.2% [Asia]; 48.4% [Europe]); exon 21 L858R substitutions (41.1% [Asia]; 29.9% [Europe]). Conclusions Although EGFR mutation prevalence was higher in Asian than Western countries, data support worldwide testing for EGFR overall and submutations to inform appropriate targeted treatment decisions. Supplementary Information The online version contains supplementary material available at 10.1007/s40291-021-00563-1.
Collapse
|
115
|
The 2021 WHO Classification of Lung Tumors: Impact of advances since 2015. J Thorac Oncol 2021; 17:362-387. [PMID: 34808341 DOI: 10.1016/j.jtho.2021.11.003] [Citation(s) in RCA: 486] [Impact Index Per Article: 162.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022]
Abstract
The 2021 World Health Organisation (WHO) Classification of Thoracic Tumours was published earlier this year, with classification of lung tumors being one of the chapters. The principles remain those of using morphology first, supported by immunohistochemistry and then molecular techniques. In 2015, there was particular emphasis on using immunohistochemistry to make classification more accurate. In 2021, there is greater emphasis throughout the book on advances in molecular pathology across all tumor types. Major features within this edition are 1) broader emphasis on genetic testing than in the 2015 WHO Classification, 2) a chapter entirely dedicated to the classification of small diagnostic samples, 3) continued recommendation to document percentages of histological patterns in invasive non-mucinous adenocarcinomas, with utilization of these features to apply a formal grading system, as well as using only invasive size for T-factor size determination in part lepidic non-mucinous lung adenocarcinomas as recommended by the 8th Edition TNM Classification, 4) recognition of spread through airspaces (STAS) as a histological feature with prognostic significance, 5) moving lymphoepithelial carcinoma to squamous cell carcinomas, 6) update on evolving concepts in lung neuroendocrine neoplasm classification, 7) recognition of bronchiolar adenoma/ciliated muconodular papillary tumor (BA/CMPT) as a new entity within the adenoma subgroup, 8) recognition of thoracic SMARCA4-deficient undifferentiated tumor, and 9) inclusion of essential and desirable diagnostic criteria for each tumor.
Collapse
|
116
|
Ericson Lindquist K, Gudinaviciene I, Mylona N, Urdar R, Lianou M, Darai-Ramqvist E, Haglund F, Béndek M, Bardoczi E, Dobra K, Brunnström H. Real-World Diagnostic Accuracy and Use of Immunohistochemical Markers in Lung Cancer Diagnostics. Biomolecules 2021; 11:1721. [PMID: 34827719 PMCID: PMC8615395 DOI: 10.3390/biom11111721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVES Accurate and reliable diagnostics are crucial as histopathological type influences selection of treatment in lung cancer. The aim of this study was to evaluate real-world accuracy and use of immunohistochemical (IHC) staining in lung cancer diagnostics. MATERIALS AND METHODS The diagnosis and used IHC stains for small specimens with lung cancer on follow-up resection were retrospectively investigated for a 15-month period at two major sites in Sweden. Additionally, 10 pathologists individually suggested diagnostic IHC staining for 15 scanned bronchial and lung biopsies and cytological specimens. RESULTS In 16 (4.7%) of 338 lung cancer cases, a discordant diagnosis of potential clinical relevance was seen between a small specimen and the follow-up resection. In half of the cases, there was a different small specimen from the same investigational work-up with a concordant diagnosis. Diagnostic inaccuracy was often related to a squamous marker not included in the IHC panel (also seen for the scanned cases), the case being a neuroendocrine tumor, thyroid transcription factor-1 (TTF-1) expression in squamous cell carcinomas (with clone SPT24), or poor differentiation. IHC was used in about 95% of cases, with a higher number of stains in biopsies and in squamous cell carcinomas and especially neuroendocrine tumors. Pre-surgical transthoracic samples were more often diagnostic than bronchoscopic ones (72-85% vs. 9-53% for prevalent types). CONCLUSIONS Although a high overall diagnostic accuracy of small specimens was seen, small changes in routine practice (such as consequent inclusion of p40 and TTF-1 clone 8G7G3/1 in the IHC panel for non-small cell cancer with unclear morphology) may lead to improvement, while reducing the number of IHC stains would be preferable from a time and cost perspective.
Collapse
Affiliation(s)
- Kajsa Ericson Lindquist
- Department of Genetics and Pathology, Laboratory Medicine Region Skåne, SE-221 85 Lund, Sweden; (K.E.L.); (I.G.); (N.M.); (R.U.); (M.L.)
- Division of Pathology, Department of Clinical Sciences, Lund University, SE-221 00 Lund, Sweden
| | - Inga Gudinaviciene
- Department of Genetics and Pathology, Laboratory Medicine Region Skåne, SE-221 85 Lund, Sweden; (K.E.L.); (I.G.); (N.M.); (R.U.); (M.L.)
| | - Nektaria Mylona
- Department of Genetics and Pathology, Laboratory Medicine Region Skåne, SE-221 85 Lund, Sweden; (K.E.L.); (I.G.); (N.M.); (R.U.); (M.L.)
| | - Rodrigo Urdar
- Department of Genetics and Pathology, Laboratory Medicine Region Skåne, SE-221 85 Lund, Sweden; (K.E.L.); (I.G.); (N.M.); (R.U.); (M.L.)
| | - Maria Lianou
- Department of Genetics and Pathology, Laboratory Medicine Region Skåne, SE-221 85 Lund, Sweden; (K.E.L.); (I.G.); (N.M.); (R.U.); (M.L.)
| | - Eva Darai-Ramqvist
- Department of Clinical Pathology and Cytology, Karolinska University Hospital Solna, SE-171 76 Stockholm, Sweden; (E.D.-R.); (F.H.)
| | - Felix Haglund
- Department of Clinical Pathology and Cytology, Karolinska University Hospital Solna, SE-171 76 Stockholm, Sweden; (E.D.-R.); (F.H.)
- Department of Oncology-Pathology, Karolinska Institute, SE-171 77 Stockholm, Sweden
| | - Mátyás Béndek
- Department of Clinical Pathology and Cytology, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden; (M.B.); (E.B.); (K.D.)
| | - Erika Bardoczi
- Department of Clinical Pathology and Cytology, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden; (M.B.); (E.B.); (K.D.)
| | - Katalin Dobra
- Department of Clinical Pathology and Cytology, Karolinska University Hospital Huddinge, SE-141 86 Stockholm, Sweden; (M.B.); (E.B.); (K.D.)
- Division of Laboratory Medicine, Department of Pathology, Karolinska Institute, SE-141 86 Stockholm, Sweden
| | - Hans Brunnström
- Department of Genetics and Pathology, Laboratory Medicine Region Skåne, SE-221 85 Lund, Sweden; (K.E.L.); (I.G.); (N.M.); (R.U.); (M.L.)
- Division of Pathology, Department of Clinical Sciences, Lund University, SE-221 00 Lund, Sweden
| |
Collapse
|
117
|
Malapelle U, Pilotto S, Reale ML, Passiglia F, Pisapia P, Pepe F, Belluomini L, Galetta D, Cortinovis D, Tiseo M, Passaro A, Seminati D, Pagni F, Parra HS, Migliorino MR, Rocco D, Troncone G, Novello S. Epidermal growth factor receptor exon 20 insertion variants in non-small cell lung cancer patients. Crit Rev Oncol Hematol 2021; 169:103536. [PMID: 34801697 DOI: 10.1016/j.critrevonc.2021.103536] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/18/2021] [Accepted: 11/15/2021] [Indexed: 12/22/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) exon 20 insertions occur rarely among different cancer types, with the highest frequency reported among non-small-cell lung cancer (NSCLC) patients, particularly adenocarcinomas (ADCs). Exon 20 insertions fall back in the tyrosine kinase domain, and can be clustered into two principal groups represented by in frame insertions and three to 21 bp (corresponding to 1-7 amino acids) duplications within amino acids 762 and 774. The identification of these alterations is key for an adequate management of NSCLC patients due to the possibility to treat these patients with specific targeted therapies. Next generation sequencing (NGS) technology, able to detect several hotspot gene mutations for different patients simultaneously, is the best detection approach due to its higher sensitivity and specificity compared to other techniques. Here we reviewed the principal biological characteristics, the main detection technologies and treatment options for NSCLC patients harbouring EGFR exon 20 insertions.
Collapse
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Sara Pilotto
- U.O.C. of Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Maria Lucia Reale
- Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Francesco Passiglia
- Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Lorenzo Belluomini
- U.O.C. of Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy
| | - Domenico Galetta
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Diego Cortinovis
- SC Oncologia Medica, SS Lung Unit Asst Ospedale San Gerardo, Monza, Italy
| | - Marcello Tiseo
- Department of Medicine and Surgery, Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Davide Seminati
- Department of Medicine and Surgery, Pathology, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Hector Soto Parra
- Department of Oncology, Medical Oncology, University Hospital Policlinico-San Marco, Catania, Italy
| | | | - Danilo Rocco
- Pneumo-Oncology Unit, Ospedali dei Colli Monaldi Cotugno CTO, Napoli, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Silvia Novello
- Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy.
| |
Collapse
|
118
|
Shokoohi A, Al-Hashami Z, Moore S, Pender A, Wong SK, Wang Y, Leung B, Wu J, Ho C. Effect of targeted therapy and immunotherapy on advanced nonsmall-cell lung cancer outcomes in the real world. Cancer Med 2021; 11:86-93. [PMID: 34786889 PMCID: PMC8704182 DOI: 10.1002/cam4.4427] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 12/26/2022] Open
Abstract
The evolution of diagnosis and treatment of advanced nonsmall‐cell lung cancer (NSCLC) has led to increasing the use of targeted therapy and immune checkpoint inhibitors. The study goal was to assess the effect of molecular testing and the introduction of new therapies on overall survival (OS). All patients with stage IV NSCLC referred to BC Cancer were included in the study. Four 1‐year time cohorts were created based on molecular testing implementation and funded drug availability: C1 baseline (2009), C2 EGFR TKI access (2011), C3 ALK inhibitor access (2015), C4 immunotherapy availability (2017). Baseline demographics, disease characteristics, and systemic therapy details were collected retrospectively. OS was calculated using the Kaplan–Meier method and compared using the log‐rank test. There were 3421 patients identified with stage IV NSCLC and 1319 (39%) received systemic therapy. In the four 1‐year time cohorts C1/C2/C3/C4: driver mutation‐targeted treatment increased 1/17/27/34% (of total systemic therapy), as did treatment with any line immunotherapy <1/1/9/38%. Median OS with best supportive care (BSC) was 3.4/3.1/3.2/2.9 m (p = 0.16) and with systemic treatment 9.9/10.9/13.9/15.0 m (p < 0.001). Median OS by treatment exposure was BSC 3.1 m, chemotherapy only 7.3 m, targeted therapy 17.5 m, and immunotherapy 20.7 m. In our real‐world study, following the introduction of targeted therapy and immune checkpoint inhibitors, there was a significant improvement in OS in each successive time cohort concordant with advancements in therapeutic options.
Collapse
Affiliation(s)
- Aria Shokoohi
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Zamzam Al-Hashami
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sara Moore
- Division of Medical Oncology, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Alexandra Pender
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Selina K Wong
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ying Wang
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bonnie Leung
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Jonn Wu
- Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Radiation Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Cheryl Ho
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
119
|
Canterbury CR, Fernandes H, Crapanzano JP, Murty VV, Mansukhani MM, Shu CA, Szabolcs M, Saqi A. ALK Gene Rearrangements in Lung Adenocarcinomas: Concordance of Immunohistochemistry, Fluorescence In Situ Hybridization, RNA In Situ Hybridization, and RNA Next-Generation Sequencing Testing. JTO Clin Res Rep 2021; 2:100223. [PMID: 34746883 PMCID: PMC8552107 DOI: 10.1016/j.jtocrr.2021.100223] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 08/05/2021] [Accepted: 08/29/2021] [Indexed: 11/29/2022] Open
Abstract
Introduction The 2018 updated molecular testing guidelines for patients with advanced lung cancer incorporated ALK immunohistochemistry (IHC) analysis as an equivalent to fluorescence in situ hybridization (FISH) method recommended in 2013. Nevertheless, no specific recommendation for alternative methods was proposed owing to insufficient data. The aim of this study was to compare the results of ALK IHC, FISH, RNA next-generation sequencing (NGS), and RNA in situ hybridization (ISH) with available clinical data. Methods A search for lung carcinomas with ALK testing by greater than or equal to one modality (i.e., ALK IHC, FISH, NGS) was performed; a subset underwent RNA ISH. When available, clinical data were recorded. Results The results were concordant among all performed testing modalities in 86 of 90 cases (95.6%). Of the four discordant cases, two were ALK positive by FISH but negative by IHC, RNA NGS, and RNA ISH. The remaining two cases failed RNA NGS testing, one was IHC negative, FISH positive, RNA ISH negative and the second was IHC positive, FISH positive, RNA ISH equivocal. RNA NGS identified one rare and one novel ALK fusion. Sufficient therapy data were available in 10 cases treated with tyrosine kinase inhibitors; three had disease progression, including one with discordant results (FISH positive, RNA NGS negative, IHC negative, RNA ISH negative) and two with concordant ALK positivity among all modalities. Conclusions Our results reveal high concordance among IHC, RNA NGS, and RNA ISH. In cases of discordance with available RNA NGS, FISH result was positive whereas IHC and ISH results were negative. On the basis of our data, multimodality testing is recommended to identify discrepant results and patients (un)likely to respond to tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Carleigh R Canterbury
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Helen Fernandes
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - John P Crapanzano
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Vundavalli V Murty
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Mahesh M Mansukhani
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Catherine A Shu
- Division of Hematology/Oncology, Department of Medicine, Columbia University Medical Center, New York, New York
| | - Matthias Szabolcs
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| | - Anjali Saqi
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| |
Collapse
|
120
|
Guirado M, Sanchez-Hernandez A, Pijuan L, Teixido C, Gómez-Caamaño A, Cilleruelo-Ramos Á. Quality indicators and excellence requirements for a multidisciplinary lung cancer tumor board by the Spanish Lung Cancer Group. Clin Transl Oncol 2021; 24:446-459. [PMID: 34665437 PMCID: PMC8525055 DOI: 10.1007/s12094-021-02712-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/18/2021] [Indexed: 12/24/2022]
Abstract
Multidisciplinary care is needed to decide the best therapeutic approach and to provide optimal care to patients with lung cancer (LC). Multidisciplinary teams (MDTs) are optimal strategies for the management of patients with LC and have been associated with better outcomes, such as an increase in quality of life and survival. The Spanish Lung Cancer Group has promoted this review about the current situation of the existing national LC-MDTs, which also offers a set of excellence requirements and quality indicators to achieve the best care in any patient with LC. Time and sufficient resources; leadership; administrative and institutional support; and recording of activity are key factors for the success of LC-MDTs. A set of excellence requirements in terms of staff, resources and organization of the LC-MDT have been proposed. At last, a list of quality indicators has been agreed to achieve and measure the performance of current LC-MDTs.
Collapse
Affiliation(s)
- M Guirado
- Medical Oncology Department, Hospital General Universitario de Elche, 03203, Elche, Spain
| | - A Sanchez-Hernandez
- Medical Oncology Department, Consorcio Hospitalario Provincial de Castellón, 12002, Castellón de la Plana, Spain
| | - L Pijuan
- Pathology Department, Bellvitge University Hospital, 08907, L'Hospitalet de Llobregat, Spain
| | - C Teixido
- Thoracic Oncology Unit, Department of Pathology, IDIBAPS, Hospital Clinic of Barcelona, C. de Villarroel, 170, 08036, Barcelona, Spain.
| | - A Gómez-Caamaño
- Department of Radiation Oncology, Hospital Clínico Universitario Santiago de Compostela, 15706, Santiago de Compostela, Spain
| | - Á Cilleruelo-Ramos
- Thoracic Surgery Department, Hospital Clínico Universitario Valladolid, 47005, Valladolid, Spain
| |
Collapse
|
121
|
Rolfo C, Mack P, Scagliotti GV, Aggarwal C, Arcila ME, Barlesi F, Bivona T, Diehn M, Dive C, Dziadziuszko R, Leighl N, Malapelle U, Mok T, Peled N, Raez LE, Sequist L, Sholl L, Swanton C, Abbosh C, Tan D, Wakelee H, Wistuba I, Bunn R, Freeman-Daily J, Wynes M, Belani C, Mitsudomi T, Gandara D. Liquid Biopsy for Advanced NSCLC: A Consensus Statement From the International Association for the Study of Lung Cancer. J Thorac Oncol 2021; 16:1647-1662. [PMID: 34246791 DOI: 10.1016/j.jtho.2021.06.017] [Citation(s) in RCA: 265] [Impact Index Per Article: 88.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/03/2021] [Accepted: 06/21/2021] [Indexed: 12/11/2022]
Abstract
Although precision medicine has had a mixed impact on the clinical management of patients with advanced-stage cancer overall, for NSCLC, and more specifically for lung adenocarcinoma, the advances have been dramatic, largely owing to the genomic complexity and growing number of druggable oncogene drivers. Furthermore, although tumor tissue is historically the "accepted standard" biospecimen for these molecular analyses, there are considerable innate limitations. Thus, liquid biopsy represents a practical alternative source for investigating tumor-derived somatic alterations. Although data are most robust in NSCLC, patients with other cancer types may also benefit from this minimally invasive approach to facilitate selection of targeted therapies. The liquid biopsy approach includes a variety of methodologies for circulating analytes. From a clinical point of view, plasma circulating tumor DNA is the most extensively studied and widely adopted alternative to tissue tumor genotyping in solid tumors, including NSCLC, first entering clinical practice for detection of EGFR mutations in NSCLC. Since the publication of the first International Association for the Study of Lung Cancer (IASLC) liquid biopsy statement in 2018, several additional advances have been made in this field, leading to changes in the therapeutic decision-making algorithm for advanced NSCLC and prompting this 2021 update. In view of the novel and impressive technological advances made in the past few years, the growing clinical application of plasma-based, next-generation sequencing, and the recent Food and Drug and Administration approval in the United States of two different assays for circulating tumor DNA analysis, IASLC revisited the role of liquid biopsy in therapeutic decision-making in a recent workshop in October 2020 and the question of "plasma first" versus "tissue first" approach toward molecular testing for advanced NSCLC. Moreover, evidence-based recommendations from IASLC provide an international perspective on when to order which test and how to interpret the results. Here, we present updates and additional considerations to the previous statement article as a consensus from a multidisciplinary and international team of experts selected by IASLC.
Collapse
Affiliation(s)
- Christian Rolfo
- Center for Thoracic Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Philip Mack
- Center for Thoracic Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Giorgio V Scagliotti
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Italy
| | - Charu Aggarwal
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maria E Arcila
- Department of Pathology, Molecular Diagnostics Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fabrice Barlesi
- CRCM, CNRS, INSERM, Aix Marseille University, Marseille, France; Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Trever Bivona
- Department of Medicine, University of California San Francisco, San Francisco, California; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California; Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Caroline Dive
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, United Kingdom; Cancer Research UK Lung Cancer Centre of Excellence, University of Manchester, Manchester, United Kingdom
| | - Rafal Dziadziuszko
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Natasha Leighl
- Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Tony Mok
- State Key Laboratory of Translational Oncology, Chinese University of Hong Kong, Hong Kong
| | - Nir Peled
- The Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka University Medical Center, Beer-Sheva, Israel
| | - Luis E Raez
- Thoracic Oncology Program, Memorial Cancer Institute/Memorial Health Care System, Florida International University, Miami, Florida
| | - Lecia Sequist
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Department of Biostatistics, Massachusetts General Hospital, Boston, Massachusetts; Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Chris Abbosh
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Daniel Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Heather Wakelee
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rebecca Bunn
- International Association for the Study of Lung Cancer, Aurora, Colorado
| | | | - Murry Wynes
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chandra Belani
- Department of Medicine Penn State College of Medicine, Penn State Cancer Institute, Hershey, Pennsylvania
| | - Tetsuya Mitsudomi
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Ohno-Higashi, Osaka-Sayama, Japan
| | - David Gandara
- Division of Hematology/Oncology, UC Davis Comprehensive Cancer Center, Sacramento, California.
| |
Collapse
|
122
|
Moore DA, Benafif S, Poskitt B, Argue S, Lee SM, Ahmad T, Papadatos-Pastos D, Jamal-Hanjani M, Bennett P, Forster MD. Optimising fusion detection through sequential DNA and RNA molecular profiling of non-small cell lung cancer. Lung Cancer 2021; 161:55-59. [PMID: 34536732 DOI: 10.1016/j.lungcan.2021.08.008] [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: 06/25/2021] [Revised: 08/15/2021] [Accepted: 08/18/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVES There is an increasing number of driver fusions in NSCLC which are amenable to targeted therapy. Panel testing for fusions is increasingly appropriate but can be costly and requires adequate good quality biopsy material. In light of the typical mutual exclusivity of driver events in NSCLC, the objective of this study was to trial a novel testing pathway, supported by industrial collaboration, in which only patients negative for driver mutations on DNA-NGS were submitted for fusion panel analysis. MATERIALS AND METHODS Over 18 months, all patients from a single centre with non-squamous NSCLC were submitted for DNA-NGS, plus ALK and ROS1 immunohistochemistry +/- FISH. Those which were negative for a driver mutation were then recalled for RNA panel testing. RESULTS 307 samples were referred for DNA-NGS mutation analysis, of which, 10% of cases were unsuitable for or failed DNA-NGS analysis. Driver mutations were detected in 61% (167/275) of all those successfully tested. Of those without a driver mutation and with some remaining tissue available, 28% had insufficient tissue/extracted RNA or failed RNA-NGS. Of those successfully tested, 24% (17/72) had a fusion gene detected involving either ALK, ROS, MET, RET, FGFR or EGFR. Overall, 66% (184/277) of patients had a driver event detected through the combination of DNA and RNA panels. CONCLUSION Sequential DNA and RNA based molecular profiling increased the efficacy of detecting fusion driven NSCLCs. Continued optimisation of tissue procurement, handling and the diagnostic pathways for gene fusion analysis is necessary to reduce analysis failure rates and improve detection rate for treatment with the next generation of small molecule inhibitors.
Collapse
Affiliation(s)
- David A Moore
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, UCL, London, United Kingdom; Department of Cellular Pathology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Sarah Benafif
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Benjamin Poskitt
- Sarah Cannon Molecular Diagnostics, HCA Healthcare UK, London, United Kingdom
| | - Stephanie Argue
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Siow-Ming Lee
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, UCL, London, United Kingdom; Department of Oncology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Tanya Ahmad
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, UCL, London, United Kingdom; Department of Oncology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Dionysis Papadatos-Pastos
- Department of Oncology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Mariam Jamal-Hanjani
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, UCL, London, United Kingdom; Department of Oncology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Philip Bennett
- Sarah Cannon Molecular Diagnostics, HCA Healthcare UK, London, United Kingdom
| | - Martin D Forster
- CRUK Lung Cancer Centre of Excellence, UCL Cancer Institute, UCL, London, United Kingdom; Department of Oncology, University College London Hospitals NHS Foundation Trust, London, United Kingdom.
| |
Collapse
|
123
|
Sunami K, Bando H, Yatabe Y, Naito Y, Takahashi H, Tsuchihara K, Toyooka S, Mimori K, Kohsaka S, Uetake H, Kinoshita I, Komine K, Takeda M, Hayashida T, Tamura K, Nishio K, Yamamoto N. Appropriate use of cancer comprehensive genome profiling assay using circulating tumor DNA. Cancer Sci 2021; 112:3911-3917. [PMID: 34128569 PMCID: PMC8409307 DOI: 10.1111/cas.15022] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 01/11/2023] Open
Abstract
Comprehensive genomic profiling (CGP) is being increasingly used for the routine clinical management of solid cancers. In July 2018, the use of tumor tissue-based CGP assays became available for all solid cancers under the universal health insurance system in Japan. Several restrictions presently exist, such as patient eligibility and limitations on the opportunities to perform such assays. The clinical implementation of CGP based on plasma circulating tumor DNA (ctDNA) is also expected to raise issues regarding the selection and use of tissue DNA and ctDNA CGP. A Joint Task Force for the Promotion of Cancer Genome Medicine comprised of three Japanese cancer-related societies has formulated a policy proposal for the appropriate use of plasma CGP (in Japanese), available at https://www.jca.gr.jp/researcher/topics/2021/files/20210120.pdf, http://www.jsco.or.jp/jpn/user_data/upload/File/20210120.pdf, and https://www.jsmo.or.jp/file/dl/newsj/2765.pdf. Based on these recommendations, the working group has summarized the respective advantages and cautions regarding the use of tissue DNA CGP and ctDNA CGP with reference to the advice of a multidisciplinary expert panel, the preferred use of plasma specimens over tissue, and multiple ctDNA testing. These recommendations have been prepared to maximize the benefits of performing CGP assays and might be applicable in other countries and regions.
Collapse
Affiliation(s)
- Kuniko Sunami
- Department of Laboratory MedicineNational Cancer Center HospitalTokyoJapan
| | - Hideaki Bando
- Department of Clinical OncologyAichi Cancer Center HospitalNagoyaJapan
| | - Yasushi Yatabe
- Department of Diagnostic PathologyNational Cancer Center HospitalDivision of Molecular PathologyNational Cancer Center Research InstituteTokyoJapan
| | - Yoichi Naito
- Department of Medical OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Hideaki Takahashi
- Department of Hepatobiliary and Pancreatic OncologyNational Cancer Center Hospital EastKashiwaJapan
| | - Katsuya Tsuchihara
- Division of Translational InformaticsExploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaJapan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrine SurgeryOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| | | | - Shinji Kohsaka
- Division of Cellular SignalingNational Cancer Center Research InstituteTokyoJapan
| | - Hiroyuki Uetake
- Department of Clinical ResearchNational Disaster Medical CenterTokyoJapan
| | - Ichiro Kinoshita
- Division of Clinical Cancer GenomicsHokkaido University HospitalSapporoJapan
| | - Keigo Komine
- Department of Clinical OncologyTohoku University HospitalSendaiJapan
| | - Masayuki Takeda
- Department of Cancer Genomics and Medical OncologyNara Medical UniversityNaraJapan
| | - Tetsu Hayashida
- Department of SurgeryKeio University School of MedicineTokyoJapan
| | - Kenji Tamura
- Department of Medical OncologyShimane University HospitalIzumoJapan
| | - Kazuto Nishio
- Department of Genome BiologyKindai University Faculty of MedicineOsakasayamaJapan
| | - Noboru Yamamoto
- Department of Experimental TherapeuticsNational Cancer Center HospitalTokyoJapan
| | | |
Collapse
|
124
|
Drenner K, Basu GD, Goodman LJ, Ozols AA, LoBello JR, Royce T, Gordon MS, Borazanci EH, Steinbach MA, Trent J, Sharma S. The value of comprehensive genomic sequencing to maximize the identification of clinically actionable alterations in advanced cancer patients: a case series. Oncotarget 2021; 12:1836-1847. [PMID: 34504655 PMCID: PMC8416559 DOI: 10.18632/oncotarget.28046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/27/2021] [Indexed: 01/10/2023] Open
Abstract
PURPOSE We present seven cases of advanced cancer patients who initially underwent tumor testing utilizing smaller, panel-based tests, followed by a variety of therapeutic treatments which ultimately resulted in progression of their disease. These cases demonstrate the value of utilizing WES/RNA seq and characterization following disease progression in these patients and the determination of clinically targetable alterations as well as acquired resistance mutations. MATERIALS AND METHODS All patients are part of an IRB approved observational study. WES and RNA sequencing were performed, using GEM ExTra® on tumor and blood samples obtained during routine clinical care. To accurately determine somatic versus germline alterations the test was performed with paired normal testing from peripheral blood. RESULTS The presented cases demonstrate the clinical impact of actionable findings uncovered using GEM ExTra® in patients with advanced disease who failed many rounds of treatment. Unique alterations were identified resulting in newly identified potential targeted therapies, mechanisms of resistance, and variation in the genomic characterization of the primary versus the metastatic tumor. CONCLUSIONS Taken together our results demonstrate that GEM ExTra® maximizes detection of actionable mutations, thus allowing for appropriate treatment selection for patients harboring both common and rare genomic alterations.
Collapse
Affiliation(s)
- Kevin Drenner
- Translational Genomic Research Institute (Tgen), Phoenix, AZ 85004, USA
- These authors contributed equally to this work
| | - Gargi D. Basu
- Ashion Analytics, LLC, Phoenix, AZ 85004, USA
- These authors contributed equally to this work
| | | | | | | | | | | | | | | | - Jeffrey Trent
- Translational Genomic Research Institute (Tgen), Phoenix, AZ 85004, USA
| | - Sunil Sharma
- Translational Genomic Research Institute (Tgen), Phoenix, AZ 85004, USA
| |
Collapse
|
125
|
Bollig-Fischer A, Bao B, Manning M, Dyson G, Michelhaugh SK, Mittal S, Bepler G, Mamdani H. Role of novel cancer gene SLITRK3 to activate NTRK3 in squamous cell lung cancer. MOLECULAR BIOMEDICINE 2021; 2:26. [PMID: 35006496 PMCID: PMC8607376 DOI: 10.1186/s43556-021-00051-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/04/2021] [Indexed: 11/10/2022] Open
Abstract
The development of targeted therapies that inhibit cancer-driving oncogenes has improved outcomes of patients diagnosed with lung adenocarcinoma (LUAD). In contrast, patients diagnosed with lung squamous cell carcinoma (LUSC) suffer worse survival outcomes and lack effective targeted treatment options. Identification of molecular drivers of LUSC to support development of targeted treatments is urgently needed. Addressing this need, the current report introduces the novel cancer gene SLIT- and NTRK-like family member 3 (SLITRK3) and its role in activating the neurotrophic receptor tyrosine kinase 3 (NTRK3) in LUSC cells. Multiple genome-wide data sets from patient samples were produced by us or downloaded from public databases to analyze tumor gene copy number aberrations, mRNA expression and associated survival outcomes. An accompanying mechanistic study employed LUSC cell lines and multiple methods, including in situ immunofluorescence, sphere-formation assay, and fluorescence-activated cell sorting analysis of the CD133-positive cell fraction. Altogether, the results indicate that gene amplification and consequent high expression of SLITRK3 in LUSC is associated with worse outcomes and induces SLITRK3-dependent activation of NTRK3 to promote a cancer stem cell phenotype that is inhibited by existing NTRK-targeted inhibitors. Based on a recent literature search, this is the first report of a mechanistic role for SLITRK3 in cancer.
Collapse
Affiliation(s)
- Aliccia Bollig-Fischer
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA.
- Department of Oncology, Wayne State University School of Medicine, 4100 John R. St., Detroit, MI, 48201, USA.
| | - Bin Bao
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
- Department of Oncology, Wayne State University School of Medicine, 4100 John R. St., Detroit, MI, 48201, USA
| | - Morenci Manning
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
- Department of Oncology, Wayne State University School of Medicine, 4100 John R. St., Detroit, MI, 48201, USA
| | - Greg Dyson
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
- Department of Oncology, Wayne State University School of Medicine, 4100 John R. St., Detroit, MI, 48201, USA
| | - Sharon K Michelhaugh
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Present Address: Carilion Clinic, Roanoke, VA, 24104, USA
- Virginia Tech Fralin Biomedical Research Institute, Roanoke, Virginia, 24016, USA
| | - Sandeep Mittal
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, 48201, USA
- Present Address: Carilion Clinic, Roanoke, VA, 24104, USA
- Virginia Tech Fralin Biomedical Research Institute, Roanoke, Virginia, 24016, USA
| | - Gerold Bepler
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
- Department of Oncology, Wayne State University School of Medicine, 4100 John R. St., Detroit, MI, 48201, USA
| | - Hirva Mamdani
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
- Department of Oncology, Wayne State University School of Medicine, 4100 John R. St., Detroit, MI, 48201, USA
| |
Collapse
|
126
|
Thai AA, Solomon BJ, Sequist LV, Gainor JF, Heist RS. Lung cancer. Lancet 2021; 398:535-554. [PMID: 34273294 DOI: 10.1016/s0140-6736(21)00312-3] [Citation(s) in RCA: 1047] [Impact Index Per Article: 349.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/14/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023]
Abstract
Lung cancer is one of the most frequently diagnosed cancers and the leading cause of cancer-related deaths worldwide with an estimated 2 million new cases and 1·76 million deaths per year. Substantial improvements in our understanding of disease biology, application of predictive biomarkers, and refinements in treatment have led to remarkable progress in the past two decades and transformed outcomes for many patients. This seminar provides an overview of advances in the screening, diagnosis, and treatment of non-small-cell lung cancer and small-cell lung cancer, with a particular focus on targeted therapies and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Alesha A Thai
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC, Australia
| | - Benjamin J Solomon
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC, Australia
| | - Lecia V Sequist
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca S Heist
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
127
|
Trosman JR, Weldon CB, Rapkin BD, Benson AB, Makower DF, Liang SY, Kulkarni SA, Perez CB, Lo SS, Krueger EA, Throckmorton AD, Gallagher C, Hoskins K, Schaeffer CM, Van Horn J, Schapira L, Ravelo A, Yu E, Gradishar WJ. Evaluation of the Novel 4R Oncology Care Planning Model in Breast Cancer: Impact on Patient Self-Management and Care Delivery in Safety-Net and Non-Safety-Net Centers. JCO Oncol Pract 2021; 17:e1202-e1214. [PMID: 34375560 DOI: 10.1200/op.21.00161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Optimal cancer care requires patient self-management and coordinated timing and sequence of interdependent care. These are challenging, especially in safety-net settings treating underserved populations. We evaluated the 4R Oncology model (4R) of patient-facing care planning for impact on self-management and delivery of interdependent care at safety-net and non-safety-net institutions. METHODS Ten institutions (five safety-net and five non-safety-net) evaluated the 4R intervention from 2017 to 2020 with patients with stage 0-III breast cancer. Data on self-management and care delivery were collected via surveys and compared between the intervention cohort and the historical cohort (diagnosed before 4R launch). 4R usefulness was assessed within the intervention cohort. RESULTS Survey response rate was 63% (422/670) in intervention and 47% (466/992) in historical cohort. 4R usefulness was reported by 79.9% of patients receiving 4R and was higher for patients in safety-net than in non-safety-net centers (87.6%, 74.2%, P = .001). The intervention cohort measured significantly higher than historical cohort in five of seven self-management metrics, including clarity of care timing and sequence (71.3%, 55%, P < .001) and ability to manage care (78.9%, 72.1%, P = .02). Referrals to interdependent care were significantly higher in the intervention than in the historical cohort along all six metrics, including primary care consult (33.9%, 27.7%, P = .045) and flu vaccination (38.6%, 27.9%, P = .001). Referral completions were significantly higher in four of six metrics. For safety-net patients, improvements in most self-management and care delivery metrics were similar or higher than for non-safety-net patients, even after controlling for all other variables. CONCLUSION 4R Oncology was useful to patients and significantly improved self-management and delivery of interdependent care, but gaps remain. Model enhancements and further evaluations are needed for broad adoption. Patients in safety-net settings benefited from 4R at similar or higher rates than non-safety-net patients, indicating that 4R may reduce care disparities.
Collapse
Affiliation(s)
- Julia R Trosman
- Center for Business Models in Healthcare, Glencoe, IL.,Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Christine B Weldon
- Center for Business Models in Healthcare, Glencoe, IL.,Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Bruce D Rapkin
- Montefiore Medical Center, Albert Einstein Cancer Center, Bronx, NY.,Department of Epidemiology and Population Health, Albert Einstein College of Medicine, New York, NY
| | - Al B Benson
- Northwestern Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | | | - Su-Ying Liang
- Sutter Health-Palo Alto Medical Foundation Research Institute, Palo Alto, CA
| | - Swati A Kulkarni
- Northwestern Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| | | | - Shelly S Lo
- Loyola University Medical Center, Maywood, IL
| | | | | | | | - Kent Hoskins
- Division of Medical Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL
| | - Cathleen M Schaeffer
- Division of Medical Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL
| | - Jennifer Van Horn
- Banner MD Anderson Cancer Center, Loveland, CO, Formerly Cheyenne Regional Medical Center, Cheyenne, WY
| | - Lidia Schapira
- Stanford University and Stanford Cancer Institute, Stanford, CA
| | | | - Elaine Yu
- Genentech Inc, South San Francisco, CA
| | - William J Gradishar
- Northwestern Medicine, Chicago, IL.,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL
| |
Collapse
|
128
|
Lee Deak K, Jackson JB, Valkenburg KC, Keefer LA, Robinson Gerding KM, Angiuoli SV, Datto MB, McCall SJ. Next-Generation Sequencing Concordance Analysis of Comprehensive Solid Tumor Profiling between a Centralized Specialty Laboratory and the Decentralized Personal Genome Diagnostics, Inc., Elio Tissue Complete Kitted Solution. J Mol Diagn 2021; 23:1324-1333. [PMID: 34314880 PMCID: PMC8567158 DOI: 10.1016/j.jmoldx.2021.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 06/11/2021] [Accepted: 07/07/2021] [Indexed: 11/09/2022] Open
Abstract
Genomic tumor profiling by next-generation sequencing (NGS) allows for large-scale tumor testing to inform targeted cancer therapies and immunotherapies, and to identify patients for clinical trials. These tests are often underutilized in patients with late-stage solid tumors and are typically performed in centralized specialty laboratories, thereby limiting access to these complex tests. Personal Genome Diagnostics Inc., elio tissue complete NGS solution is a comprehensive DNA-to-report kitted assay and bioinformatics solution. Comparison of 147 unique specimens from >20 tumor types was performed using the elio tissue complete solution and Foundation Medicine's FoundationOne test, which is of similar size and gene content. The analytical performance of all genomic variant types was evaluated. In general, the overall mutational profile is highly concordant between the two assays, with agreement in sequence variants reported between panels demonstrating >95% positive percentage agreement for single-nucleotide variants and insertions/deletions in clinically actionable genes. Both copy number alterations and gene translocations showed 80% to 83% positive percentage agreement, whereas tumor mutation burden and microsatellite status showed a high level of concordance across a range of mutation loads and tumor types. The Personal Genome Diagnostics Inc., elio tissue complete assay is comparable to the FoundationOne test and will allow more laboratories to offer a diagnostic NGS assay in house, which will ultimately reduce time to result and increase the number of patients receiving molecular genomic profiling and personalized treatment.
Collapse
Affiliation(s)
- Kristen Lee Deak
- Department of Pathology, Duke University Medical Center, Durham, North Carolina.
| | | | | | | | | | | | - Michael B Datto
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Shannon J McCall
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
129
|
Dziadziuszko R, Mok T, Peters S, Han JY, Alatorre-Alexander J, Leighl N, Sriuranpong V, Pérol M, de Castro Junior G, Nadal E, de Marinis F, Frontera OA, Tan DSW, Lee DH, Kim HR, Yan M, Riehl T, Schleifman E, Paul SM, Mocci S, Patel R, Assaf ZJ, Shames DS, Mathisen MS, Gadgeel SM. Blood First Assay Screening Trial (BFAST) in Treatment-Naive Advanced or Metastatic NSCLC: Initial Results of the Phase 2 ALK-Positive Cohort. J Thorac Oncol 2021; 16:2040-2050. [PMID: 34311110 DOI: 10.1016/j.jtho.2021.07.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/02/2021] [Accepted: 07/04/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION The Blood First Assay Screening Trial is an ongoing open-label, multicohort study, prospectively evaluating the relationship between blood-based next-generation sequencing (NGS) detection of actionable genetic alterations and activity of targeted therapies or immunotherapy in treatment-naive advanced or metastatic NSCLC. We present data from the ALK-positive cohort. METHODS Patients aged more than or equal to 18 years with stage IIIB or IV NSCLC and ALK rearrangements detected by blood-based NGS using hybrid capture technology (FoundationACT) received alectinib 600 mg twice daily. Asymptomatic or treated central nervous system (CNS) metastases were permitted. Primary end point was investigator-assessed objective response rate (ORR; Response Evaluation Criteria in Solid Tumors version 1.1). Secondary end points were independent review facility-assessed ORR, duration of response, progression-free survival (PFS), overall survival, and safety. Exploratory end points were investigator-assessed ORR in patients with baseline CNS metastases and relationship between circulating biomarkers and response. RESULTS In total, 2219 patients were screened and blood-based NGS yielded results in 98.6% of the cases. Of these, 119 patients (5.4%) had ALK-positive disease; 87 were enrolled and received alectinib. Median follow-up was 12.6 months (range: 2.6-18.7). Confirmed ORR was 87.4% (95% confidence interval [CI]: 78.5-93.5) by investigator and 92.0% (95% CI: 84.1-96.7) by independent review facility. Investigator-confirmed 12-month duration of response was 75.9% (95% CI: 63.6-88.2). In 35 patients (40%) with baseline CNS disease, investigator-assessed ORR was 91.4% (95% CI: 76.9-98.2). Median PFS was not reached; 12-month investigator-assessed PFS was 78.4% (95% CI: 69.1-87.7). Safety data were consistent with the known tolerability profile of alectinib. CONCLUSIONS These results reveal the clinical application of blood-based NGS as a method to inform clinical decision-making in ALK-positive NSCLC.
Collapse
Affiliation(s)
- Rafal Dziadziuszko
- Department of Oncology and Radiotherapy, Medical University of Gdansk, Gdansk, Poland
| | - Tony Mok
- State Key Laboratory of Translational Oncology, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Solange Peters
- Oncology Department, University Hospital (CHUV), University of Lausanne, Switzerland
| | - Ji-Youn Han
- Center for Lung Cancer, National Cancer Center, Goyang, South Korea
| | | | - Natasha Leighl
- Division of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Virote Sriuranpong
- Faculty of Medicine, Chulalongkorn University and the King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Maurice Pérol
- Department of Medical Oncology, Léon Bérard Cancer Center, Lyon, France
| | | | - Ernest Nadal
- Catalan Institute of Oncology, L'Hospitalet, Barcelona, Spain
| | - Filippo de Marinis
- European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | | | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Dae Ho Lee
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Centre, Yonsei University College of Medicine, Seoul, South Korea
| | - Mark Yan
- F. Hoffmann-La Roche, Mississauga, Canada
| | - Todd Riehl
- Genentech, Inc., South San Francisco, California
| | | | - Sarah M Paul
- Genentech, Inc., South San Francisco, California
| | | | - Rajesh Patel
- Genentech, Inc., South San Francisco, California
| | | | | | | | - Shirish M Gadgeel
- Department of Internal Medicine, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, Michigan.
| |
Collapse
|
130
|
Nakra T, Singh V, Nambirajan A, Malik PS, Mohan A, Jain D. Correlation of TTF-1 immunoexpression and EGFR mutation spectrum in non-small cell lung carcinoma. J Pathol Transl Med 2021; 55:279-288. [PMID: 34233113 PMCID: PMC8353134 DOI: 10.4132/jptm.2021.05.10] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/10/2021] [Indexed: 01/18/2023] Open
Abstract
Background Thyroid transcription factor (TTF-1) is a diagnostic marker expressed in 75%–85% of primary lung adenocarcinomas (ACs). Activating mutations in the tyrosine kinase domain of the epidermal growth factor receptor (EGFR) gene is the most common targetable driver alteration in lung AC. Previous studies have shown a positive correlation between TTF-1 and EGFR mutation status. We aimed to determine the predictive value of TTF-1 immunoexpression for underlying EGFR mutation status in a large Indian cohort. Methods This retrospective designed study was conducted with medical record data from 2011 to 2020. All cases of primary lung AC and non–small cell lung carcinoma not otherwise specified (NSCLC, NOS) with known TTF-1 expression diagnosed by immunohistochemistry using 8G7G3/1 antibodies and EGFR mutation status diagnosed by quantitative polymerase chain reaction were retrieved, reviewed, and theresults were analyzed. Results Among 909 patient samples diagnosed as lung AC and NSCLC, NOS, TTF-1 was positive in 76.8% cases (698/909) and EGFR mutations were detected in 29.6% (269/909). A strong positive correlation was present between TTF-1 positivity and EGFR mutation status (odds ratio, 3.61; p < .001), with TTF-1 positivity showing high sensitivity (90%) and negative predictive value (87%) for EGFR mutation. TTF-1 immunoexpression did not show significant correlation with uncommon/dual EGFR mutations (odds ratio, 1.69; p = .098). EGFR–tyrosine kinase inhibitor therapy was significantly superior to chemotherapy among EGFR mutant cases irrespective of TTF-1 status; however, no significant differences among survival outcomes were observed. Conclusions Our study confirms a strong positive correlation between TTF-1 expression and common EGFR mutations (exon 19 deletion and exon 21 L858R) in advanced lung AC with significantly high negative predictive value of TTF-1 for EGFR mutations.
Collapse
Affiliation(s)
- Tripti Nakra
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Varsha Singh
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Aruna Nambirajan
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Prabhat Singh Malik
- Department of Medical Oncology, Dr B.R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Anant Mohan
- Department of Pulmonary Medicine and Sleep Disorders, All India Institute of Medical Sciences, New Delhi, India
| | - Deepali Jain
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
131
|
Abstract
The identification of targetable genomic alterations in lung cancer is required as standard of care to guide optimal therapy selection. With a constantly evolving landscape of ancillary molecular and biomarker testing in lung cancer, pathologists need to be aware of what specimens to test, how the testing should be performed, and which targets to test for to provide the clinically relevant genomic information necessary to treat these patients. Several guideline statements on the topic are currently available to help pathologists and laboratory personnel best use the small specimens obtained from patients with lung cancer for ancillary molecular testing.
Collapse
Affiliation(s)
- Sinchita Roy-Chowdhuri
- Department of Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard Unit 83, Houston, TX 77030, USA.
| |
Collapse
|
132
|
Status quo of ALK testing in lung cancer: results of an EQA scheme based on in-situ hybridization, immunohistochemistry, and RNA/DNA sequencing. Virchows Arch 2021; 479:247-255. [PMID: 34173019 PMCID: PMC8364534 DOI: 10.1007/s00428-021-03106-5] [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: 09/22/2020] [Revised: 04/07/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022]
Abstract
With this external quality assessment (EQA) scheme, we aim to investigate the diagnostic performance of the currently available methods for the detection of ALK alterations in non-small cell lung cancer on a national scale, namely, in situ hybridization (ISH), immunohistochemistry (IHC), and RNA/DNA sequencing (NGS). The EQA scheme cohort consisted of ten specimens, including four ALK positive and six ALK negative samples, which were thoroughly pretested using IHC, ISH, and RNA/DNA NGS. Unstained tumor sections were provided to the 57 participants, and the results were retrieved via an online questionnaire. ISH was used by 29, IHC by 38, and RNA/DNA sequencing by 19 participants. Twenty-eight institutions (97%) passed the ring trial using ISH, 33 (87%) by using IHC, and 18 (95%) by using NGS. The highest sensitivity and interrater agreement (Fleiss ‘ kappa) was observed for RNA/DNA sequencing (99%, 0.975), followed by ISH (94%, 0.898) and IHC (92%, 0.888). However, the proportion of samples that were not evaluable due to bad tissue quality was also higher for RNA/DNA sequencing (4%) compared with ISH (0.7%) and IHC (0.5%). While all three methods produced reliable results between the different institutions, the highest sensitivity and concordance were observed for RNA/DNA sequencing. These findings encourage the broad implementation of this method in routine diagnostic, although the application might be limited by technical capacity, economical restrictions, and tissue quality of formalin-fixed samples.
Collapse
|
133
|
Wittlinger F, Laufer SA. The pre-clinical discovery and development of osimertinib used to treat non-small cell lung cancer. Expert Opin Drug Discov 2021; 16:1091-1103. [PMID: 34053372 DOI: 10.1080/17460441.2021.1936496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Osimertinib is currently the only FDA- and EMA-approved third-generation small-molecule epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI). It was initially indicated for second-line treatment of patients with metastatic EGFR T790M mutation-positive non-small cell lung cancer (NSCLC) and got approved for first-line treatment of EGFR activation mutation-positive metastatic NSCLC in 2018. Most recently, the FDA granted approval for the adjuvant treatment of patients with early-stage mutated EGFR NSCLC after tumor resection.Areas covered: This drug discovery case history focuses on the key studies that led to the preclinical discovery and development of osimertinib. The authors focus on published preclinical studies by scientists from AstraZeneca and highlight key events in the clinical development.Expert opinion: Although eventually compromised by the cellular plasticity of the tumor and the inevitable acquisition of drug resistance through the use of osimertinib, its key role in the treatment of NSCLC with specific EGFR mutations will be maintained in the near future. As the genome of EGFR is highly labile and since the rapid development of new mutants remains an issue, there is still room for improvement for the next generation of inhibitors.
Collapse
Affiliation(s)
- Florian Wittlinger
- Tuebingen Center for Academic Drug Discovery & Development, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Stefan A Laufer
- Tuebingen Center for Academic Drug Discovery & Development, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Tübingen, Germany
| |
Collapse
|
134
|
Comparison of Resistance Spectra after First and Second Line Osimertinib Treatment Detected by Liquid Biopsy. Cancers (Basel) 2021; 13:cancers13122861. [PMID: 34201252 PMCID: PMC8227553 DOI: 10.3390/cancers13122861] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Since the recent approval of osimertinib, a third generation tyrosine kinase inhibitor (TKI) targeting EGFR in non-small cell lung cancer (NSCLC), tracing the resistance mechanisms that yield to failure of osimertinib has become of interest. As the spectrum of osimertinib-resistance related genomic alterations appears significantly more diverse compared to first and second generation TKI, comprehensive, and preferably non-invasive molecular diagnostic methods are required for the detection of resistance mechanisms. In this study, we present molecular results of 56 NSCLC patients during disease progression on first and second line osimertinib treatment using a hybrid capture (HC) next generation sequencing (NGS) based liquid biopsy approach. We show examples of polyclonal resistance development which leads to the presence of multiple resistance mechanisms in the same patient, and highlight the clinical utility of HC NGS over single gene testing. Abstract Since 2009, several first, second, and third generation EGFR tyrosine kinase inhibitors (TKI) have been approved for targeted treatment of EGFR mutated metastatic non-small lung cancer (NSCLC). A vast majority of patients is improving quickly on treatment; however, resistance is inevitable and typically occurs after one year for TKI of the first and second generation. Osimertinib, a third generation TKI, has recently been approved for first line treatment in the palliative setting and is expected to become approved for the adjuvant setting as well. Progression-free survival (PFS) under osimertinib is superior to its predecessors but its spectrum of resistance alterations appears significantly more diverse compared to first and second generation EGFR TKI. As resistance mechanisms to osimertinib are therapeutically targetable in some cases, it is important to comprehensively test for molecular alterations in the relapse scenario. Liquid biopsy may be advantageous over tissue analysis as it has the potential to represent tumor heterogeneity and clonal diversification. We have previously shown high concordance of hybrid capture (HC) based next generation sequencing (NGS) in liquid biopsy versus solid tumor biopsies. In this study, we now present real-word data from 56 patients with metastatic NSCLC that were tested by liquid biopsy at the time of disease progression on mostly second line treated osimertinib treatment. We present examples of single and multiple TKI resistance mechanisms, including mutations in multiple pathways, copy number changes and rare fusions of RET, ALK, FGFR3 and BRAF. In addition, we present the added value of HC based NGS to reveal polyclonal resistance development at the DNA level encoding multiple EGFR C797S and PIK3CA mutations.
Collapse
|
135
|
Cuppens K, Lodewyckx L, Demedts I, Decoster L, Colinet B, Deschepper K, Janssens A, Galdermans D, Pieters T. Real-World Treatment Patterns, Epidermal Growth Factor Receptor (EGFR) Testing and Outcomes in EGFR-Mutated Advanced Non-small Cell Lung Cancer Patients in Belgium: Results from the REVEAL Study. Drugs Real World Outcomes 2021; 8:141-152. [PMID: 33710523 PMCID: PMC8128953 DOI: 10.1007/s40801-021-00243-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Treatment of patients with epidermal growth factor receptor-mutated (EGFRm) non-small cell lung cancer (NSCLC) continues to evolve expeditiously. OBJECTIVES This retrospective study investigated real-world treatment patterns and EGFR mutation testing in patients with EGFRm advanced NSCLC in Belgium. METHODS Data were extracted from medical records of adults diagnosed with EGFRm locally advanced/metastatic NSCLC between 1 September 2015 and 31 December 2017. Patients were followed retrospectively from diagnosis until 1 September 2018, end of clinical activity or death. Data on demographics, patient outcomes and disease characteristics, treatment patterns and EGFR mutation testing at diagnosis and progression were analyzed descriptively. RESULTS A total of 141 patients were enrolled. At diagnosis, median age was 69 years, 63.1% were female, 88.7% had metastatic disease, 94.3% had adenocarcinoma histology, 76.6% had ECOG 0/1, 70.9% had common EGFR mutations and 29.1% had only rare mutations. In first line, 73.8% of patients received first/second-generation EGFR-tyrosine kinase inhibitors (1G/2G EGFR-TKIs), while 21.9% received other systemic treatments. Among 61 patients progressing on and discontinuing a first 1G/2G EGFR-TKI, 45 (73.8%) received subsequent systemic treatment while 16 (26.2%) did not; 20 (32.8%) received osimertinib. Among 65 patients progressing on a first 1G/2G EGFR-TKI, 47 (72.3%) were tested for T790M, of whom 25 (53.2%) were positive. CONCLUSION These real-world data from Belgium show that a substantial fraction of patients with EGFRm NSCLC do not receive 1G/2G EGFR-TKIs in first line and do not receive subsequent systemic treatment after progression on 1G/2G EGFR-TKIs. Only a third receive osimertinib upon progression on 1G/2G EGFR-TKIs. These observations should be considered in first-line treatment decisions. TRIAL REGISTRATION ClinicalTrials.gov: NCT03761901-December 3, 2018.
Collapse
Affiliation(s)
- Kristof Cuppens
- Department of Pulmonology and Thoracic Oncology, Jessa Hospital, Stadsomvaart 11, 3500, Hasselt, Belgium.
| | | | - Ingel Demedts
- Department of Pulmonary Diseases, AZ Delta, Roeselare, Belgium
| | - Lore Decoster
- Department of Medical Oncology, Oncologisch Centrum, Vrije Universiteit Brussel, BrusselJette, UZ, Belgium
| | - Benoît Colinet
- Department of Pneumology and Thoracic Oncology, Grand Hôpital de Charleroi, Notre Dame, Charleroi, Belgium
| | | | - Annelies Janssens
- Department of Pulmonology and Thoracic Oncology, Antwerp University Hospital, Edegem, Belgium
| | | | - Thierry Pieters
- Department of Pulmonology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
136
|
Seet AOL, Tan AC, Tan TJ, Ng MCH, Tai DWM, Lam JYC, Tan GS, Gogna A, Too CW, Tan BS, Takano A, Lim A, Lim TH, Lim ST, Dent RA, Ang MK, Yap YS, Tan IBH, Choo SP, Toh CK, Lim EH, Farid M, Skanderup AJ, Iyer NG, Lim WT, Tan EH, Lim TKH, Tan DSW. Individualized Molecular Profiling for Allocation to Clinical Trials Singapore Study-An Asian Tertiary Cancer Center Experience. JCO Precis Oncol 2021; 5:PO.20.00261. [PMID: 34250396 DOI: 10.1200/po.20.00261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/02/2020] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Precision oncology has transformed the management of advanced cancers through implementation of advanced molecular profiling technologies to identify increasingly defined subsets of patients and match them to appropriate therapy. We report outcomes of a prospective molecular profiling study in a high-volume Asian tertiary cancer center. PATIENTS AND METHODS Patients with advanced cancer were enrolled onto a prospective protocol for genomic profiling, the Individualized Molecular Profiling for Allocation to Clinical Trials Singapore study, at the National Cancer Center Singapore. Primary objective was to identify molecular biomarkers in patient's tumors for allocation to clinical trials. The study commenced in February 2012 and is ongoing, with the results of all patients who underwent multiplex next-generation sequencing (NGS) testing until December 2018 presented here. The results were discussed at a molecular tumor board where recommendations for allocation to biomarker-directed trials or targeted therapies were made. RESULTS One thousand fifteen patients were enrolled with a median age of 58 years (range 20-83 years). Most common tumor types were lung adenocarcinoma (26%), colorectal cancer (15%), and breast cancer (12%). A total of 1,064 NGS assays were performed, on fresh tumor tissue for 369 (35%) and archival tumor tissue for 687 (65%) assays. TP53 (39%) alterations were most common, followed by EGFR (21%), KRAS (14%), and PIK3CA (10%). Of 405 NGS assays with potentially actionable alterations, 111 (27%) were allocated to a clinical trial after molecular tumor board and 20 (4.9%) were enrolled on a molecularly matched clinical trial. Gene fusions were detected in 23 of 311 (7%) patients tested, including rare fusions in new tumor types and known fusions in rare tumors. CONCLUSION Individualized Molecular Profiling for Allocation to Clinical Trials Singapore demonstrates the feasibility of a prospective broad molecular profiling program in an Asian tertiary cancer center, with the ability to develop and adapt to a dynamic landscape of precision oncology.
Collapse
Affiliation(s)
- Amanda O L Seet
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Aaron C Tan
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Tira J Tan
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Matthew C H Ng
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - David W M Tai
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Justina Y C Lam
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Gek San Tan
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Apoorva Gogna
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Singapore, Singapore
| | - Chow Wei Too
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Singapore, Singapore
| | - Bien Soo Tan
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Singapore, Singapore
| | - Angela Takano
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Alvin Lim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Tse Hui Lim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Soon Thye Lim
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | | | - Mei Kim Ang
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Yoon-Sim Yap
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Iain B H Tan
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Su Pin Choo
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Chee Keong Toh
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Elaine H Lim
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Mohamad Farid
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | | | - N Gopalakrishna Iyer
- Division of Surgical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Wan Teck Lim
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Eng Huat Tan
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Tony K H Lim
- Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
137
|
Strohmeier S, Brcic I, Popper H, Liegl-Atzwanger B, Lindenmann J, Brcic L. Applicability of pan-TRK immunohistochemistry for identification of NTRK fusions in lung carcinoma. Sci Rep 2021; 11:9785. [PMID: 33963267 PMCID: PMC8105314 DOI: 10.1038/s41598-021-89373-3] [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: 03/16/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
In the last two decades, various therapies have been introduced for lung carcinoma patients, including tyrosine-kinase inhibitors for different mutations. While some of them are specific to specific tumor types, others, like NTRK1-3 fusions, are found in various solid tumors. The occurrence of an NTRK1,2 or 3 fusion acts as a biomarker for efficient treatment with NTRK inhibitors, irrespectively of the tumor type. However, the occurrence of the NTRK1-3 fusions in lung carcinomas is extremely rare. We performed a retrospective analysis to evaluate the applicability of immunohistochemistry with the pan-TRK antibody in the detection of NTRK fusions in lung carcinomas. The study cohort included 176 adenocarcinomas (AC), 161 squamous cell carcinomas (SCC), 31 large-cell neuroendocrine carcinomas (LCNEC), and 19 small cell lung carcinomas (SCLC). Immunohistochemistry (IHC) was performed using the pan-TRK antibody (clone EPR17341, Ventana) on tissue microarrays, while confirmation for all positive cases was done using RNA-based Archer FusionPlex MUG Lung Panel. On IHC staining, 12/387 samples (3.1%) demonstrated a positive reaction. Ten SCC cases (10/161, 6.2%), and two LCNEC cases (2/31, 6.5%) were positive. Positive cases demonstrated heterogeneous staining of tumor cells, mostly membranous with some cytoplasmic and in one case nuclear pattern. RNA-based sequencing did not demonstrate any NTRK1-3 fusion in our patients' collective. Our study demonstrates that pan-TRK expression in lung carcinoma is very low across different histologic types. NTRK1-3 fusions using an RNA-based sequencing approached could not be detected. This stresses the importance of confirmation of immunohistochemistry results by molecular methods.
Collapse
Affiliation(s)
- Simon Strohmeier
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010, Graz, Austria
| | - Iva Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010, Graz, Austria
| | - Helmut Popper
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010, Graz, Austria
| | - Bernadette Liegl-Atzwanger
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010, Graz, Austria
| | - Jörg Lindenmann
- Division of Thoracic Surgery and Hyperbaric Surgery, Department of Surgery, Medical University of Graz, Auenbruggerplatz 29/3, 8036, Graz, Austria
| | - Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010, Graz, Austria.
| |
Collapse
|
138
|
Conde E, Hernandez S, Benito A, Caminoa A, Garrido P, Lopez-Rios F. Screening for ROS1 fusions in patients with advanced non-small cell lung carcinomas using the VENTANA ROS1 (SP384) Rabbit Monoclonal Primary Antibody. Expert Rev Mol Diagn 2021; 21:437-444. [PMID: 33899645 DOI: 10.1080/14737159.2021.1919512] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: The development of several ROS1 inhibitors means that the importance of accurately identifying ROS1-positive lung cancer patients has never been greater. Therefore, it is crucial that ROS1 testing assays become more standardized.Areas covered: Based on primary literature, combined with personal diagnostic and research experience, this review provide a pragmatic update on the use of the recently released VENTANA ROS1 (SP384) Rabbit Monoclonal Primary Antibody.Expert opinion: This assay provides high sensitivity, so it is an excellent analytical option when screening for ROS1 fusions in patients with advanced non-small cell lung carcinomas.
Collapse
Affiliation(s)
- Esther Conde
- Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HM Hospitales, CIBERONC, Madrid, Spain
| | - Susana Hernandez
- Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HM Hospitales, Madrid, Spain
| | - Amparo Benito
- Pathology, Ramon Y Cajal University Hospital, Madrid, Spain
| | | | - Pilar Garrido
- Medical Oncology, Ramon Y Cajal University Hospital, CIBERONC, Madrid, Spain
| | - Fernando Lopez-Rios
- Pathology and Laboratory of Therapeutic Targets, Hospital Universitario HM Sanchinarro, HM Hospitales, CIBERONC, Madrid, Spain
| |
Collapse
|
139
|
Li W, Li Y, Guo L, Liu Y, Yang L, Ying J. Metastatic NSCLCs With Limited Tissues: How to Effectively Identify Driver Alterations to Guide Targeted Therapy in Chinese Patients. JTO Clin Res Rep 2021; 2:100167. [PMID: 34590019 PMCID: PMC8474491 DOI: 10.1016/j.jtocrr.2021.100167] [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: 12/19/2020] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Molecular diagnostics of newly diagnosed patients with metastatic NSCLC (mNSCLC) with limited tissue samples often face several obstacles in routine practice using next-generation sequencing (NGS), mainly owing to insufficient tissue or DNA; thus, how to effectively identify the molecular profiling of these cases to accurately guide targeted therapy remains elusive. We evaluated whether an optimized workflow with the combined use of multiple technologies could be helpful. METHODS Tissue NGS was used as the frontline method. Amplification refractory mutation system polymerase chain reaction, immunohistochemistry, fluorescence in situ hybridization, and plasma NGS were used as supplements. RESULTS Among 208 mNSCLC cases with limited tissue (cohort 1), molecular genotyping using single-tissue NGS failed in 42 (20.2%) and actionable alterations were identified in only 112 of 208 cases (53.8%). In comparison, the optimized workflow in 1184 additional mNSCLC cases with limited tissue (cohort 2) increased the discovery rate of actionable alterations from 59.7% detected by tissue NGS to 70.4%. It was because that driver alterations were identified using amplification refractory mutation system polymerase chain reaction plus immunohistochemistry or fluorescence in situ hybridization in 53 of 78 (67.9%) tissue NGS-failed cases, and using plasma NGS in 73 of 143 (51.0%) tissue NGS-failed cases, which led to matched targeted therapies in 57 cases with clinical response. Moreover, the median turnaround time of the optimized workflow was significantly shorter than that of repeated biopsy for tissue NGS (p < 0.001). CONCLUSIONS The optimized workflow can improve mutation detection and may avoid repeated biopsy, thus allowing the timely initiation of targeted therapies for patients with newly diagnosed mNSCLC.
Collapse
Affiliation(s)
- Weihua Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yan Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Lin Yang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| |
Collapse
|
140
|
Hofman P. Next-Generation Sequencing with Liquid Biopsies from Treatment-Naïve Non-Small Cell Lung Carcinoma Patients. Cancers (Basel) 2021; 13:2049. [PMID: 33922637 PMCID: PMC8122958 DOI: 10.3390/cancers13092049] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/11/2021] [Accepted: 04/20/2021] [Indexed: 12/16/2022] Open
Abstract
Recently, the liquid biopsy (LB), a non-invasive and easy to repeat approach, has started to compete with the tissue biopsy (TB) for detection of targets for administration of therapeutic strategies for patients with advanced stages of lung cancer at tumor progression. A LB at diagnosis of late stage non-small cell lung carcinoma (NSCLC) is also being performed. It may be asked if a LB can be complementary (according to the clinical presentation or systematics) or even an alternative to a TB for treatment-naïve advanced NSCLC patients. Nucleic acid analysis with a TB by next-generation sequencing (NGS) is gradually replacing targeted sequencing methods for assessment of genomic alterations in lung cancer patients with tumor progression, but also at baseline. However, LB is still not often used in daily practice for NGS. This review addresses different aspects relating to the use of LB for NGS at diagnosis in advanced NSCLC, including its advantages and limitations.
Collapse
Affiliation(s)
- Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Université Côte d’Azur, CHU Nice, FHU OncoAge, Pasteur Hospital, 30 avenue de la voie romaine, BP69, CEDEX 01, 06001 Nice, France; ; Tel.: +33-4-92-03-88-55 or +33-4-92-03-87-49; Fax: +33-4-92-88-50
- Hospital-Integrated Biobank BB-0033-00025, Université Côte d’Azur, CHU Nice, FHU OncoAge, 06001 Nice, France
| |
Collapse
|
141
|
Zhang T, Xu Z, Liu G, Jiang B, de Bock GH, Groen HJM, Vliegenthart R, Xie X. Simultaneous Identification of EGFR,KRAS,ERBB2, and TP53 Mutations in Patients with Non-Small Cell Lung Cancer by Machine Learning-Derived Three-Dimensional Radiomics. Cancers (Basel) 2021; 13:cancers13081814. [PMID: 33920322 PMCID: PMC8070114 DOI: 10.3390/cancers13081814] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary Multiple genetic mutations are associated with the outcomes of patients with non-small cell lung cancer (NSCLC) after using tyrosine kinase inhibitor, but the cost for detecting multiple genetic mutations is high. Few studies have investigated whether multiple genetic mutations can be simultaneously detected based on image features in patients with NSCLC. We developed a machine learning-derived radiomics approach that can simultaneously discriminate the presence of EGFR, KRAS, ERBB2, and TP53 mutations on CT images in patients with NSCLC. These findings suggest that machine learning-derived radiomics may become a noninvasive and low-cost method to screen for multiple genetic mutations in patients with NSCLC before using next-generation sequencing tests, which can help to improve individualized targeted therapies. Abstract Purpose: To develop a machine learning-derived radiomics approach to simultaneously discriminate epidermal growth factor receptor (EGFR), Kirsten rat sarcoma viral oncogene (KRAS), Erb-B2 receptor tyrosine kinase 2 (ERBB2), and tumor protein 53 (TP53) genetic mutations in patients with non-small cell lung cancer (NSCLC). Methods: This study included consecutive patients from April 2018 to June 2020 who had histologically confirmed NSCLC, and underwent pre-surgical contrast-enhanced CT and post-surgical next-generation sequencing (NGS) tests to determine the presence of EGFR, KRAS, ERBB2, and TP53 mutations. A dedicated radiomics analysis package extracted 1672 radiomic features in three dimensions. Discriminative models were established using the least absolute shrinkage and selection operator to determine the presence of EGFR, KRAS, ERBB2, and TP53 mutations, based on radiomic features and relevant clinical factors. Results: In 134 patients (63.6 ± 8.9 years), the 20 most relevant radiomic features (13 for KRAS) to mutations were selected to construct models. The areas under the curve (AUCs) of the combined model (radiomic features and relevant clinical factors) for discriminating EGFR, KRAS, ERBB2, and TP53 mutations were 0.78 (95% CI: 0.70–0.86), 0.81 (0.69–0.93), 0.87 (0.78–0.95), and 0.84 (0.78–0.91), respectively. In particular, the specificity to exclude EGFR mutations was 0.96 (0.87–0.99). The sensitivity to determine KRAS, ERBB2, and TP53 mutations ranged from 0.82 (0.69–90) to 0.92 (0.62–0.99). Conclusions: Machine learning-derived 3D radiomics can simultaneously discriminate the presence of EGFR, KRAS, ERBB2, and TP53 mutations in patients with NSCLC. This noninvasive and low-cost approach may be helpful in screening patients before invasive sampling and NGS testing.
Collapse
Affiliation(s)
- Tiening Zhang
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China;
| | - Zhihan Xu
- Siemens Healthineers Ltd., Zhouzhu Rd.278, Shanghai 200120, China;
| | - Guixue Liu
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China; (G.L.); (B.J.)
| | - Beibei Jiang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China; (G.L.); (B.J.)
| | - Geertruida H. de Bock
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
| | - Harry J. M. Groen
- Department of Lung Diseases, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands;
| | - Rozemarijn Vliegenthart
- Department of Radiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands;
| | - Xueqian Xie
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China; (G.L.); (B.J.)
- Correspondence:
| |
Collapse
|
142
|
Park S, Olsen S, Ku BM, Lee MS, Jung HA, Sun JM, Lee SH, Ahn JS, Park K, Choi YL, Ahn MJ. High concordance of actionable genomic alterations identified between circulating tumor DNA-based and tissue-based next-generation sequencing testing in advanced non-small cell lung cancer: The Korean Lung Liquid Versus Invasive Biopsy Program. Cancer 2021; 127:3019-3028. [PMID: 33826761 DOI: 10.1002/cncr.33571] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Because of the growing number of actionable biomarkers in non-small cell lung cancer (NSCLC), sufficient tissue availability for testing is becoming a greater challenge. Liquid biopsy offers a potential solution by complementing standard tissue-based methods. In this study, the authors analyzed the concordance of actionable genomic alterations sequenced from circulating tumor DNA (ctDNA; Guardant360) and tissue (Oncomine Focus Assay). METHODS From September 2015 to May 2018, 421 paired plasma and tissue samples from patients with advanced NSCLC who had previously undergone tissue testing by standard methods were collected. Both types of samples were available for 287 patients (262 in cohort 1 [treatment-naive] and 25 in cohort 2 [treatment failure]), and only 1 sample type was available for 134 patients (50 in cohort 3 [plasma only] and 84 in cohort 4 [tissue only]). RESULTS In cohort 1, 198 samples (77.6%) showed concordance between tissue and plasma next-generation sequencing (NGS). Among the discordant cases, plasma testing detected additional genomic alterations in 11 patients (4.2%). In 50 patients without tissue-based NGS results (cohort 3), the ctDNA-based test detected genomic alterations in 20 samples (40.0%). The median allele frequency (AF) of mutations identified with ctDNA-based NGS (0.74%) was lower than that identified with the tissue-based NGS test (13.90%). Clinical responses to matched targeted therapy occurred, regardless of the ctDNA AF. Upfront ctDNA-based testing identified 60.4% of patients with genomic alterations. In addition, ctDNA-based testing uncovered 12.0% more actionable alterations when it was performed after tissue-based NGS testing. CONCLUSIONS The results indicate that a ctDNA-based test identifies additional patients with actionable genomic alterations and could, therefore, be used to complement traditional tissue-based testing for NSCLC. LAY SUMMARY Circulating tumor DNA (ctDNA)-based next-generation sequencing (NGS) testing is becoming essential as the number of actionable genomic biomarker increases for the treatment selection of non-small cell lung cancer. This study demonstrates the additive value of ctDNA-based testing in addition to tissue-based NGS and standard of care-based biomarker testing for detecting additional patients with actionable genomic alterations. Clinical responses have also been observed in patients with a low allele frequency detected by ctDNA-based NGS testing.
Collapse
Affiliation(s)
- Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Steve Olsen
- Department of Medical and Clinical Affairs, Guardant Health AMEA, Singapore, Singapore
| | - Bo Mi Ku
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Min-Sang Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun-Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Keunchil Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
143
|
Li W, Lyu Y, Wang S, Zhou X, Ma J, Xu C, Fang L, Ying J. Trends in Molecular Testing of Lung Cancer in Mainland People's Republic of China Over the Decade 2010 to 2019. JTO Clin Res Rep 2021; 2:100163. [PMID: 34590013 PMCID: PMC8474368 DOI: 10.1016/j.jtocrr.2021.100163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Lung cancer is the leading cause of cancer-related morbidity and mortality in the People's Republic of China. Targeted therapies for patients with lung cancer, which depend on accurate identification of actionable genomic alteration, have improved survival compared with previously available treatments. However, data on the types of molecular testing often used in the People's Republic of China, and how they have changed over time, are scarce. We explored the overall landscape of molecular testing of lung cancer in mainland People's Republic of China in the past decade. METHODS We distributed a stratified random sampling survey of molecular testing to 49 hospitals from members of the Molecular Pathology Collaboration Group of Chinese Anti-Cancer Association which was weighted by the numbers of lung cancer cases in seven different geographic regions in mainland People's Republic of China from 2010 to 2019. The questionnaire contained four parts for all respondents. The questionnaire ascertained the use of approved in vitro diagnostic (IVD) devices published by the Center for Medical Device Evaluation, National Medical Products Administration of the People's Republic of China. RESULTS A total of 226,227 NSCLC specimens were tested from 2010 to 2019 in the selected hospitals. The annual number of initiated molecular tests increased over time (p < 0.0001), with an average annual growth rate of 31.8%. A notable increase in the number of molecular tests occurred during 2014 and 2016, which coincided with the approval of the National Medical Products Administration to IVD devices. For the diagnosis of molecular subtypes, EGFR mutation testing was first conducted in year 2007, followed by ALK translocation testing in 2010 and ROS1 in 2011. For other rare genetic variations in NSCLC, BRAF mutation testing was first launched in 2012, MET exon 14 skipping mutation in 2014, HER2 exon 20 mutations in 2017, and RET translocation in 2015. A markedly uneven distribution was also observed in the geography of leading units with the largest number of leading units located in east People's Republic of China (34.7%, 17 of 49) and the smallest number located in northwest People's Republic of China (6.1%, 3 of 49). The growth trends we observed illustrate the progress and increasing capability of molecular testing of lung cancer achieved in mainland People's Republic of China in the decade from 2010. CONCLUSIONS In the decade 2010 to 2019, progress and increased capability of molecular testing of lung cancer were achieved in mainland People's Republic of China. Further efforts should address the clinical application of next-generation sequencing technology, rare genomic aberrations, and the balance between novel genomic testing techniques and the approval of IVD products.
Collapse
Affiliation(s)
- Wenbin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yunfeng Lyu
- Department of Clinical Evaluation and Biometrics Division II, Center for Medical Device Evaluation, National Medical Products Administration, Beijing, People’s Republic of China
| | - Shaoming Wang
- National Central Cancer Registry Office, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Xiaoyan Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
| | - Jie Ma
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, People’s Republic of China
| | - Chao Xu
- Department of Clinical Evaluation and Biometrics Division II, Center for Medical Device Evaluation, National Medical Products Administration, Beijing, People’s Republic of China
| | - Li Fang
- Department of Clinical Evaluation and Biometrics Division II, Center for Medical Device Evaluation, National Medical Products Administration, Beijing, People’s Republic of China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| |
Collapse
|
144
|
Abstract
Lung cancer is the leading cause of cancer deaths worldwide, and patients with nonsmall cell lung cancer have traditionally had a poor prognosis. An improved understanding of targetable oncogenic molecular alterations has led to a growing number of effective and first-line therapies in targeted patient populations. This review provides an overview of systemic therapy options available for patients with mutation-driven nonsmall cell lung cancer, as well as a discussion of data regarding safety when combined with radiation therapy.
Collapse
Affiliation(s)
- Lova Sun
- Division of Hematology/Oncology, Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA.
| | - Melina E Marmarelis
- Division of Hematology/Oncology, Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| | - Corey J Langer
- Division of Hematology/Oncology, Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
145
|
Tan AC, Chan J, Khasraw M. The role of immunotherapy in fusion-driven lung cancer. Expert Rev Anticancer Ther 2021; 21:461-464. [PMID: 33682589 DOI: 10.1080/14737140.2021.1899816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Aaron C Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Johan Chan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Mustafa Khasraw
- Duke Center for Cancer Immunotherapy, Duke University, Durham, NC, USA
| |
Collapse
|
146
|
Zhang G, Zhao Z, Cao Y, Zhang J, Li S, Deng L, Zhou J. Relationship between epidermal growth factor receptor mutations and CT features in patients with lung adenocarcinoma. Clin Radiol 2021; 76:473.e17-473.e24. [PMID: 33731263 DOI: 10.1016/j.crad.2021.02.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 02/05/2021] [Indexed: 01/04/2023]
Abstract
AIM The purpose of this study was to investigate the relationship between epidermal growth factor receptor (EGFR) mutation status and computed tomography (CT) features in patients with lung adenocarcinoma. MATERIALS AND METHODS A total of 483 patients with lung adenocarcinoma diagnosed between January 2015 and April 2020 were included in this study. All patients underwent a preoperative chest CT, and a total of 31 detailed CT features were quantified. The mutation status of EGFR exon 18-21 was detected by a polymerase chain reaction (PCR)-based amplified refractory mutation system. Student's t and Fisher's exact or chi-square tests were used to compare continuous and categorical variables, respectively. Least absolute shrinkage and selection operator (LASSO) regularisation was used to determine the optimal combination of CT features and clinical characteristics to predict the EGFR mutation status. The model was tested using a validation set consisting of 120 patients. RESULTS EGFR mutations were found in 249 (51.6%) of 483 patients with lung adenocarcinoma. Univariate analysis showed that 14 CT features and two clinical characteristics correlated significantly with the EGFR mutation status. Smoking history, long-axis diameter, bubble-like lucency, pleural retraction, thickened bronchovascular bundles, and peripheral emphysema were independent predictors of the EGFR mutation status, according to LASSO regularisation. In the training and verification cohorts, the areas under the curve of the prediction model were 0.766 and 0.745, respectively. CONCLUSIONS CT features of patients with lung adenocarcinoma can help predict the EGFR mutation status.
Collapse
Affiliation(s)
- G Zhang
- Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging, Gansu Province, China; Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Z Zhao
- Second Clinical School, Lanzhou University, Lanzhou, China; Department of Neurosurgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Y Cao
- Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging, Gansu Province, China
| | - J Zhang
- Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging, Gansu Province, China
| | - S Li
- Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging, Gansu Province, China
| | - L Deng
- Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging, Gansu Province, China
| | - J Zhou
- Key Laboratory of Medical Imaging, Gansu Province, China; Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
147
|
Current treatment and future challenges in ROS1- and ALK-rearranged advanced non-small cell lung cancer. Cancer Treat Rev 2021; 95:102178. [PMID: 33743408 DOI: 10.1016/j.ctrv.2021.102178] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
Non─small cell lung cancer (NSCLC) presents different druggable genetic abnormalities, including ROS1 and ALK rearrangements, which share relevant clinical features and therapeutic strategies. The homology between the tyrosine kinase domains of ROS1 and ALK defines unique subsets of patients highly sensitive to targeted tyrosine kinase inhibitors (TKIs). Genomic profiling in advanced NSCLC is standard, immunohistochemistry and fluorescence in situ hybridization being the main techniques used to detect genomic rearrangements. Personalized treatment with TKIs in ROS1- and ALK-positive NSCLC patients has dramatically improved patients' outcomes. Crizotinib has been the first-line standard of care treatment in ALK-rearranged NSCLC patients for a long time, while crizotinib still represents the best upfront therapeutic option in ROS1-positive NSCLC patients, followed by next-generation TKIs at the time of disease progression. However, the improved intracranial efficacy of next-generation TKIs has led to these drugs becoming first-line options, widening treatment opportunities for these patients. Since all patients will develop disease progression under TKI therapy, understanding the mechanisms of acquired resistance is crucial to define the optimal sequential therapeutic strategy. Despite the positive correlation between personalized treatment and patients' outcome, access to next-generation TKIs and genomic profiling at the time of disease progression are major challenges to achieving this goal. In this review, we present updated evidence on ROS1- and ALK-rearranged NSCLC regarding epidemiology and diagnostics, current therapies and the most suitable sequential treatment approaches, as well as mechanisms of acquired resistance and strategies to overcome them.
Collapse
|
148
|
Song Z, Lu C, Xu CW, Zheng Z. Noncanonical Gene Fusions Detected at the DNA Level Necessitate Orthogonal Diagnosis Methods Before Targeted Therapy. J Thorac Oncol 2021; 16:344-348. [PMID: 33641715 DOI: 10.1016/j.jtho.2020.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Zhengbo Song
- Department of Clinical Trial, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, People's Republic of China; Institute of Basic Medicine and Cancer(IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, People's Republic of China
| | - Chenyu Lu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, People's Republic of China; Biotechnology and Health Centre, City University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China
| | - Chun-Wei Xu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Zongli Zheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, People's Republic of China; Biotechnology and Health Centre, City University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China; Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong, People's Republic of China.
| |
Collapse
|
149
|
Santarpia M, Massafra M, Gebbia V, D’Aquino A, Garipoli C, Altavilla G, Rosell R. A narrative review of MET inhibitors in non-small cell lung cancer with MET exon 14 skipping mutations. Transl Lung Cancer Res 2021; 10:1536-1556. [PMID: 33889528 PMCID: PMC8044480 DOI: 10.21037/tlcr-20-1113] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Treatment of advanced non-small cell lung cancer (NSCLC) has radically improved in the last years due to development and clinical approval of highly effective agents including immune checkpoint inhibitors (ICIs) and oncogene-directed therapies. Molecular profiling of lung cancer samples for activated oncogenes, including epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), c-ros oncogene 1 (ROS1) and BRAF, is routinely performed to select the most appropriate up-front treatment. However, the identification of new therapeutic targets remains a high priority. Recently, MET exon 14 skipping mutations have emerged as novel actionable oncogenic alterations in NSCLC, sensitive to MET inhibition. In this review we discuss: (I) MET gene and MET receptor structure and signaling pathway; (II) MET exon 14 alterations; (III) current data on MET inhibitors, mainly focusing on selective MET tyrosine kinase inhibitors (TKIs), in the treatment of NSCLC with MET exon 14 skipping mutations. We identified the references for this review through a literature search of papers about MET, MET exon 14 skipping mutations, and MET inhibitors, published up to September 2020, by using PubMed, Scopus and Web of Science databases. We also searched on websites of main international cancer congresses (ASCO, ESMO, IASLC) for ongoing studies presented as abstracts. MET exon 14 skipping mutations have been associated with clinical activity of selective MET inhibitors, including capmatinib, that has recently received approval by FDA for clinical use in this subgroup of NSCLC patients. A large number of trials are testing MET inhibitors, also in combinatorial therapeutic strategies, in MET exon 14-altered NSCLC. Results from these trials are eagerly awaited to definitively establish the role and setting for use of these agents in NSCLC patients.
Collapse
Affiliation(s)
- Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Patology “G. Barresi”, University of Messina, Messina, Italy
| | - Marco Massafra
- Medical Oncology Unit, Department of Human Patology “G. Barresi”, University of Messina, Messina, Italy
| | - Vittorio Gebbia
- Medical Oncology and Supportive Care Unit, La Maddalena Cancer Center, Palermo, Italy;,Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Antonio D’Aquino
- Medical Oncology Unit, Department of Human Patology “G. Barresi”, University of Messina, Messina, Italy
| | - Claudia Garipoli
- Medical Oncology Unit, Department of Human Patology “G. Barresi”, University of Messina, Messina, Italy
| | - Giuseppe Altavilla
- Medical Oncology Unit, Department of Human Patology “G. Barresi”, University of Messina, Messina, Italy
| | - Rafael Rosell
- Catalan Institute of Oncology, Laboratory of Cellular and Molecular Biology, Institute for Health Science Research Germans Trias i Pujol, Badalona, Barcelona, Spain;,Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Dexeus University Hospital, Barcelona, Spain;,Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
150
|
The evolving landscape of biomarker testing for non-small cell lung cancer in Europe. Lung Cancer 2021; 154:161-175. [PMID: 33690091 DOI: 10.1016/j.lungcan.2021.02.026] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/18/2021] [Accepted: 02/21/2021] [Indexed: 12/31/2022]
Abstract
The discovery of oncogenic driver mutations rendering non-small cell lung cancer (NSCLC) targetable by small-molecule inhibitors, and the development of immunotherapies, have revolutionised NSCLC treatment. Today, instead of non-selective chemotherapies, all patients with advanced NSCLC eligible for treatment (and increasing numbers with earlier, less extensive disease) require fast and comprehensive screening of biomarkers for first-line patient selection for targeted therapy, chemotherapy, or immunotherapy (with or without chemotherapy). To avoid unnecessary re-biopsies, biomarker screening before first-line treatment should also include markers that are actionable from second-line onwards; PD-L1 expression testing is also mandatory before initiating treatment. Population differences exist in the frequency of oncogenic driver mutations: EGFR mutations are more frequent in Asia than Europe, whereas the converse is true for KRAS mutations. In addition to approved first-line therapies, a number of emerging therapies are being investigated in clinical trials. Guidelines for biomarker testing vary by country, with the number of actionable targets and the requirement for extensive molecular screening strategies expected to increase. To meet diagnostic demands, rapid screening technologies for single-driver mutations have been implemented. Improvements in DNA- and RNA-based next-generation sequencing technologies enable analysis of a group of genes in one assay; however, turnaround times remain relatively long. Consequently, rapid screening technologies are being implemented alongside next-generation sequencing. Further challenges in the evolving landscape of biomarker testing in NSCLC are actionable primary and secondary resistance mechanisms to targeted therapies. Therefore, comprehensive testing on re-biopsies, collected at the time of disease progression, in combination with testing of circulating tumour DNA may provide important information to guide second- or third-line therapies. Furthermore, longitudinal biomarker testing can provide insights into tumour evolution and heterogeneity during the course of the disease. We summarise best practice strategies for Europe in the changing landscape of biomarker testing at diagnosis and during treatment.
Collapse
|