1
|
Lee S, Roh J, Park JS, Tuncay IO, Lee W, Kim JA, Oh BBL, Shin JY, Lee JS, Ju YS, Kim R, Park S, Koo J, Park H, Lim J, Connolly-Strong E, Kim TH, Choi YW, Ahn MS, Lee HW, Kim S, Kim JH, Kwon M. Target-Enhanced Whole-Genome Sequencing Shows Clinical Validity Equivalent to Commercially Available Targeted Oncology Panel. Cancer Res Treat 2025; 57:350-361. [PMID: 39300929 PMCID: PMC12016826 DOI: 10.4143/crt.2024.114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024] Open
Abstract
PURPOSE Cancer poses a significant global health challenge, demanding precise genomic testing for individualized treatment strategies. Targeted-panel sequencing (TPS) has improved personalized oncology but often lacks comprehensive coverage of crucial cancer alterations. Whole-genome sequencing (WGS) addresses this gap, offering extensive genomic testing. This study demonstrates the medical potential of WGS. MATERIALS AND METHODS This study evaluates target-enhanced WGS (TE-WGS), a clinical-grade WGS method sequencing both cancer and matched normal tissues. Forty-nine patients with various solid cancer types underwent both TE-WGS and TruSight Oncology 500 (TSO500), one of the mainstream TPS approaches. RESULTS TE-WGS detected all variants reported by TSO500 (100%, 498/498). A high correlation in variant allele fractions was observed between TE-WGS and TSO500 (r=0.978). Notably, 223 variants (44.8%) within the common set were discerned exclusively by TE-WGS in peripheral blood, suggesting their germline origin. Conversely, the remaining subset of 275 variants (55.2%) were not detected in peripheral blood using the TE-WGS, signifying them as bona fide somatic variants. Further, TE-WGS provided accurate copy number profiles, fusion genes, microsatellite instability, and homologous recombination deficiency scores, which were essential for clinical decision-making. CONCLUSION TE-WGS is a comprehensive approach in personalized oncology, matching TSO500's key biomarker detection capabilities. It uniquely identifies germline variants and genomic instability markers, offering additional clinical actions. Its adaptability and cost-effectiveness underscore its clinical utility, making TE-WGS a valuable tool in personalized cancer treatment.
Collapse
Affiliation(s)
| | - Jin Roh
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Jun Sung Park
- Department of Internal Medicine, Ajou University School of Medicine, Suwon, Korea
| | | | | | | | | | | | | | | | - Ryul Kim
- Inocras Inc., San Diego, CA, USA
| | | | | | | | | | | | - Tae-Hwan Kim
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Korea
| | - Yong Won Choi
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Korea
| | - Mi Sun Ahn
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Korea
| | - Hyun Woo Lee
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Korea
| | - Seokhwi Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Jang-Hee Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Minsuk Kwon
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Korea
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
2
|
Fox AH, Osarogiagbon RU, Farjah F, Jett JR, Johnson BE, Rivera MP, Smith RA, Wistuba II, Silvestri GA. The American Cancer Society National Lung Cancer Roundtable strategic plan: Advancing comprehensive biomarker testing in non-small cell lung cancer. Cancer 2024; 130:4188-4199. [PMID: 39347617 PMCID: PMC11585345 DOI: 10.1002/cncr.34628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/05/2022] [Accepted: 04/05/2022] [Indexed: 10/01/2024]
Abstract
Comprehensive biomarker testing is a crucial requirement for the optimal treatment of advanced-stage non-small cell lung cancer (NSCLC), with emerging relevance in the adjuvant treatment setting. To advance its goal of ensuring optimal therapy for persons diagnosed with lung cancer, the American Cancer Society National Lung Cancer Roundtable (ACS NLCRT) held The Summit on Optimizing Lung Cancer Biomarkers in Practice in September 2020 to align its partners toward the goal of ensuring comprehensive biomarker testing for all eligible patients with NSCLC. The ACS NLCRT's Strategic Plan for Advancing Comprehensive Biomarker Testing in NSCLC, a product of the summit, comprises actions to promote comprehensive biomarker testing for all eligible patients. The approach is multifaceted, including policy-level advocacy and the development and dissemination of targeted educational materials, clinical decision tools, and guides to patients, physicians, and payers aimed at ameliorating barriers to testing experienced by each of these groups. PLAIN LANGUAGE SUMMARY: The ACS NLCRT works to improve care for patients with lung cancer. The ACS NLCRT supports comprehensive biomarker testing as essential to determine treatment options for all eligible patients with non-small cell lung cancer. Many factors lead to some patients not receiving optimal biomarker testing. The ACS NLCRT held a collaborative summit and developed a strategic plan to achieve and promote comprehensive biomarker testing for all patients. These plans include developing educational materials and physician tools and advocating for national policies in support of biomarker testing.
Collapse
Affiliation(s)
- Adam H. Fox
- Division of Pulmonary and Critical Care MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | | | - Farhood Farjah
- Department of SurgeryUniversity of WashingtonSeattleWashingtonUSA
| | | | - Bruce E. Johnson
- Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMassachusettsUSA
| | - M. Patricia Rivera
- Department of MedicineDivision of Pulmonary and Critical Care MedicineWilmot Cancer InstituteThe University of Rochester Medical CenterRochesterNew YorkUSA
| | - Robert A. Smith
- Center for Early Cancer Detection ScienceAmerican Cancer SocietyAtlantaGeorgiaUSA
| | - Ignacio I. Wistuba
- Department of Translational Molecular PathologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Gerard A. Silvestri
- Division of Pulmonary and Critical Care MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| |
Collapse
|
3
|
Shah A, Apple J, Burgos G, Lankin J, Cohn J, Mulvihill E, Cambron-Mellott MJ. Physician preferences of biomarker testing strategies in newly diagnosed stage IV non-small cell lung cancer patients. Future Oncol 2024; 20:3229-3243. [PMID: 39552591 DOI: 10.1080/14796694.2024.2419351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 10/17/2024] [Indexed: 11/19/2024] Open
Abstract
Aim: To understand physicians' attitudes and behaviors regarding EGFR testing and retesting strategies in newly diagnosed metastatic non-small cell lung cancer patients.Materials & methods: Oncologists and pathologists completed an online, cross-sectional survey.Results: Most oncologists (73.3%) and pathologists (53.4%) agreed that concurrent testing increases sensitivity for detecting EGFR mutations. Upon tissue insufficiency, oncologists and pathologists reported using liquid biopsy 77.0% and 39.0% of the time, respectively. Tumor accessibility, smoking status, patient willingness and age were key drivers of tissue re-biopsy. Most oncologists reported high confidence in proceeding to first-line therapy based solely on liquid biopsy (60.7-80.0%); fewer pathologists (37.9%) were comfortable with this decision.Conclusion: Variation in physicians' perceptions of testing and retesting highlights the need for greater stakeholder consensus.
Collapse
Affiliation(s)
- Anne Shah
- AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Jon Apple
- AstraZeneca, One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Gabriela Burgos
- Oracle Life Sciences, Oracle Corporation, World Headquarters, 2300 Oracle Way, Austin, TX 78741, USA
| | - Josh Lankin
- Oracle Life Sciences, Oracle Corporation, World Headquarters, 2300 Oracle Way, Austin, TX 78741, USA
| | - Jesse Cohn
- Oracle Life Sciences, Oracle Corporation, World Headquarters, 2300 Oracle Way, Austin, TX 78741, USA
| | - Emily Mulvihill
- Oracle Life Sciences, Oracle Corporation, World Headquarters, 2300 Oracle Way, Austin, TX 78741, USA
| | - M Janelle Cambron-Mellott
- Oracle Life Sciences, Oracle Corporation, World Headquarters, 2300 Oracle Way, Austin, TX 78741, USA
| |
Collapse
|
4
|
Ouchaoui AA, Hadad SEE, Aherkou M, Fadoua E, Mouad M, Ramli Y, Kettani A, Bourais I. Unlocking Benzosampangine's Potential: A Computational Approach to Investigating, Its Role as a PD-L1 Inhibitor in Tumor Immune Evasion via Molecular Docking, Dynamic Simulation, and ADMET Profiling. Bioinform Biol Insights 2024; 18:11779322241298591. [PMID: 39564188 PMCID: PMC11574905 DOI: 10.1177/11779322241298591] [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: 04/08/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
The interaction between programmed cell death protein 1 (PD-1) and its ligand PD-L1 plays a crucial role in tumor immune evasion, presenting a critical target for cancer immunotherapy. Despite being effective, current monoclonal antibodies present some drawbacks such as high costs, toxicity, and resistance development. Therefore, the development of small-molecule inhibitors is necessary, especially those derived from natural sources. In this study, benzosampangine is predicted as a promising PD-L1 inhibitor, with potential applications in cancer immunotherapy. Utilizing the high-resolution crystal structure of human PD-L1 (PDB ID: 5O45), we screened 511 natural compounds, identifying benzosampangine as a top candidate with exceptional inhibitory properties. Molecular docking predicted that benzosampangine exhibits a strong binding affinity for PD-L1 (-9.4 kcal/mol) compared with established controls such as CA-170 (-6.5 kcal/mol), BMS-202 (-8.6 kcal/mol), and pyrvinium (-8.9 kcal/mol). The compound's predicted binding efficacy is highlighted by robust interactions with key amino acids (ILE54, TYR56, GLN66, MET115, ILE116, SER117, ALA121, ASP122) within the active site, notably forming 3 Pi-sulfur interactions with MET115-an interaction absents in control inhibitors. In addition, ADMET profiling suggests that over the control molecules, benzosampangine has several key advantages, including favorable solubility, permeability, metabolic stability, and low toxicity, while adhering to Lipinski's rule of five. Molecular dynamic simulations predict the stability of the benzosampangine-PD-L1 complex, reinforcing its potential to sustain inhibition of the PD-1/PD-L1 pathway. MMGBSA analysis calculated a binding free energy (ΔGbind) of -39.39 kcal/mol for the benzosampangine-PD-L1 complex, with significant contributions from Coulombic, lipophilic, and Van der Waals interactions, validating the predicted docking results. This study investigates in silico benzosampangine, predicting its better molecular interactions and pharmacokinetic profile compared with several already known PD-L1 inhibitors.
Collapse
Affiliation(s)
- Abderrahim Ait Ouchaoui
- Mohammed VI University of Sciences and Health (UM6SS), Casablanca, Morocco
- Mohammed VI Center for Research and Innovation (CM6RI), Rabat, Morocco
| | - Salah Eddine El Hadad
- Mohammed VI University of Sciences and Health (UM6SS), Casablanca, Morocco
- Mohammed VI Center for Research and Innovation (CM6RI), Rabat, Morocco
| | - Marouane Aherkou
- Mohammed VI Center for Research and Innovation (CM6RI), Rabat, Morocco
- Biotechnology Laboratory (MedBiotech), Bioinova Research Center, Rabat Medical and Pharmacy School, Mohammed V University in Rabat, Rabat, Morocco
| | - Elkamili Fadoua
- Rabat Medical and Pharmacy School, Mohammed Vth University, Rabat, Morocco
| | - Mkamel Mouad
- Mohammed VI Center for Research and Innovation (CM6RI), Rabat, Morocco
| | - Youssef Ramli
- Laboratory of Medicinal Chemistry, Drug Sciences Research Center, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Anass Kettani
- Laboratory of Biology and Health, URAC 34, Faculty of Sciences Ben M'sik, Health and Biotechnology Research Center, Hassan II University of Casablanca, Casablanca, Morocco
| | - Ilhame Bourais
- Mohammed VI University of Sciences and Health (UM6SS), Casablanca, Morocco
- Mohammed VI Center for Research and Innovation (CM6RI), Rabat, Morocco
- Laboratory of Human Pathologies Biology, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| |
Collapse
|
5
|
Chavanel B, Virard F, Cahais V, Renard C, Sirand C, Smits KM, Schouten LJ, Fervers B, Charbotel B, Abedi-Ardekani B, Korenjak M, Zavadil J. Genome-scale mutational signature analysis in archived fixed tissues. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2024; 794:108512. [PMID: 39216514 DOI: 10.1016/j.mrrev.2024.108512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/25/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Mutation spectra and mutational signatures in cancerous and non-cancerous tissues can be identified by various established techniques of massively parallel sequencing (or next-generation sequencing) including whole-exome or whole-genome sequencing, and more recently by error-corrected/duplex sequencing. One rather underexplored area has been the genome-scale analysis of mutational signatures as markers of mutagenic exposures, and their impact on cancer driver events applied to formalin-fixed or alcohol-fixed paraffin embedded archived biospecimens. This review showcases successful applications of the next-generation sequencing methodologies in archived fixed tissues, including the delineation of the specific tissue fixation-related DNA damage manifesting as artifactual signatures, distinguishable from the true signatures that arise from biological mutagenic processes. Overall, we discuss and demonstrate how next-generation sequencing techniques applied to archived fixed biospecimens can enhance our understanding of cancer causes including mutagenic effects of extrinsic cancer risk agents, and the implications for prevention efforts aimed at reducing avoidable cancer-causing exposures.
Collapse
Affiliation(s)
- Bérénice Chavanel
- International Agency for Research on Cancer, Epigenomics and Mechanisms Branch, Lyon, France
| | - François Virard
- International Agency for Research on Cancer, Epigenomics and Mechanisms Branch, Lyon, France; University Claude Bernard Lyon 1 INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Lyon, France
| | - Vincent Cahais
- International Agency for Research on Cancer, Epigenomics and Mechanisms Branch, Lyon, France
| | - Claire Renard
- International Agency for Research on Cancer, Epigenomics and Mechanisms Branch, Lyon, France
| | - Cécilia Sirand
- International Agency for Research on Cancer, Epigenomics and Mechanisms Branch, Lyon, France
| | - Kim M Smits
- Maastricht University, Research Institute for Oncology and Reproduction, Department of Pathology, Maastricht, the Netherlands
| | - Leo J Schouten
- Maastricht University, Research Institute for Oncology and Reproduction, Department of Epidemiology, Maastricht, the Netherlands
| | - Béatrice Fervers
- Centre Léon Bérard, Department Cancer and Environment, Lyon, France
| | - Barbara Charbotel
- University Claude Bernard Lyon 1, UMRESTTE, Epidemiological Research and Surveillance Unit in Transport, Occupation and Environment, Lyon, France
| | | | - Michael Korenjak
- International Agency for Research on Cancer, Epigenomics and Mechanisms Branch, Lyon, France
| | - Jiri Zavadil
- International Agency for Research on Cancer, Epigenomics and Mechanisms Branch, Lyon, France.
| |
Collapse
|
6
|
Sheehan KN, Khoury LM, Niehaus AG, Mariencheck WI, Gershner KA, Dotson TL, Bellinger CR. Endobronchial Ultrasound Guided Transbronchial Needle Aspiration and Next Generation Sequencing Yields. Lung 2024; 202:317-324. [PMID: 38687384 PMCID: PMC11143046 DOI: 10.1007/s00408-024-00690-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/23/2024] [Indexed: 05/02/2024]
Abstract
PURPOSE The use of endobronchial ultrasound (EBUS) is standard practice for lung cancer diagnosis and staging. Next generation sequencing (NGS) for detection of genetic alterations is recommended in advanced, non-squamous, non-small-cell lung cancer (NSCLC). Existing protocols for NGS testing are minimal and reported yields vary. This study aimed to determine the yield of EBUS samples obtained for NGS using a sampling protocol at our institution and assess predictive factors to form collection protocols. METHODS We reviewed EBUS bronchoscopies from 2016 to 2021 with non-squamous NSCLC diagnoses. For target lesions suspected to be malignant, the sampling protocol was: (a) two slides for on-site evaluation, (b) three to five fine needle aspirations rinsed into saline for immunohistochemical staining and in-house molecular markers, and (c) additional three to five rinses for NGS. Sufficiency for NGS processing was determined by the pathology department. RESULTS Two hundred and seventy-eight non-squamous NSCLC samples were obtained by EBUS (205 adenocarcinoma; 73 not otherwise specified). EBUS was performed under general anesthesia in 75.5% of cases. The overall sample adequacy for NGS testing was 57.5%. Higher adequacy rates were observed when protocol was adhered to 66.0% versus 37.2% (p < 0.001). There was no statistically significant difference based on the size of the lesion or location of the sample. CONCLUSION When a protocol of three to five dedicated needle rinses for NGS was followed, we nearly doubled our sample adequacy rate for NSG as compared to standard care. Studies are needed to determine the ideal collection and processing modality to preserve tissue samples for genetic sequencing.
Collapse
Affiliation(s)
- Kristin N Sheehan
- Department of Pulmonary/Critical Care, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| | - Lara M Khoury
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Angela G Niehaus
- Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - William I Mariencheck
- Department of Pulmonary/Critical Care, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Katherine A Gershner
- Department of Pulmonary/Critical Care, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Travis L Dotson
- Department of Pulmonary/Critical Care, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Christina R Bellinger
- Department of Pulmonary/Critical Care, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| |
Collapse
|
7
|
Ferguson S, Sriram S, Wallace JK, Lee J, Kim JA, Lee Y, Oh BBL, Lee WC, Lee S, Connolly-Strong E. Analytical and Clinical Validation of a Target-Enhanced Whole Genome Sequencing-Based Comprehensive Genomic Profiling Test. Cancer Invest 2024; 42:390-399. [PMID: 38773925 DOI: 10.1080/07357907.2024.2352438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/03/2024] [Indexed: 05/24/2024]
Abstract
Evaluation of the test performance of the Target enhanced whole-genome sequencing (TE-WGS) assay for comprehensive oncology genomic profiling. The analytical validation of the assay included sensitivity and specificity for single nucleotide variants (SNVs), insertions/deletions (indels), and structural variants (SVs), revealing a revealed a sensitivity of 99.8% for SNVs and 99.2% for indels. The positive predictive value (PPV) was 99.3% SNVs and 98.7% indels. Clinical validation was benchmarked against established orthogonal methods and demonstrated high concordance with reference methods. TE-WGS provides insights beyond targeted panels by comprehensive analysis of key biomarkers and the entire genome encompassing both germline and somatic findings.
Collapse
|
8
|
Jansen JP, Ragavan MV, Chen C, Douglas MP, Phillips KA. The Health Inequality Impact of Liquid Biopsy to Inform First-Line Treatment of Advanced Non-Small Cell Lung Cancer: A Distributional Cost-Effectiveness Analysis. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:1697-1710. [PMID: 37741446 PMCID: PMC10859998 DOI: 10.1016/j.jval.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/28/2023] [Accepted: 08/10/2023] [Indexed: 09/25/2023]
Abstract
OBJECTIVES To perform a distributional cost-effectiveness analysis of liquid biopsy (LB) followed by, if needed, tissue biopsy (TB) (LB-first strategy) relative to a TB-only strategy to inform first-line treatment of advanced non-small cell lung cancer (aNSCLC) from a US payer perspective by which we quantify the impact of LB-first on population health inequality according to race and ethnicity. METHODS With a health economic model, quality-adjusted life-years (QALYs) and costs per patient were estimated for each subgroup. Given the lifetime risk of aNSCLC, and assuming equally distributed opportunity costs, the incremental net health benefits of LB-first were calculated, which were used to estimate general population quality-adjusted life expectancy at birth (QALE) by race and ethnicity with and without LB-first. The degree of QALYs and QALE differences with the strategies was expressed with inequality indices. Their differences were defined as the inequality impact of LB-first. RESULTS LB-first resulted in an additional 0.21 (95% uncertainty interval: 0.07-0.39) QALYs among treated patients, with the greatest gain observed among Asian patients (0.31 QALYs [0.09-0.61]). LB-first resulted in an increase in relative inequality in QALYs among patients, but a minor decrease in relative inequality in QALE. CONCLUSIONS LB-first to inform first-line aNSCLC therapy can improve health outcomes. With current diagnostic performance, the benefit is the greatest among Asian patients, thereby potentially widening racial and ethnic differences in survival among patients with aNSCLC. Assuming equally distributed opportunity costs and access, LB-first does not worsen and, in fact, may reduce inequality in general population health according to race and ethnicity.
Collapse
Affiliation(s)
- Jeroen P Jansen
- Department of Clinical Pharmacy, UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA, USA; UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA; UCSF Philip R. Lee Institute for Health Policy, San Francisco, CA, USA.
| | - Meera V Ragavan
- Division of Hematology and Oncology, UCSF Department of Medicine, San Francisco, CA, USA
| | - Cheng Chen
- Department of Clinical Pharmacy, UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA, USA
| | - Michael P Douglas
- Department of Clinical Pharmacy, UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA, USA
| | - Kathryn A Phillips
- Department of Clinical Pharmacy, UCSF Center for Translational and Policy Research on Precision Medicine (TRANSPERS), San Francisco, CA, USA; UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA; UCSF Philip R. Lee Institute for Health Policy, San Francisco, CA, USA
| |
Collapse
|
9
|
Wakuda K, Yamaguchi H, Kenmotsu H, Fukuda M, Ito K, Tsuchiya-Kawano Y, Tanaka K, Harada T, Nakatani Y, Miura S, Yokoyama T, Nakamura T, Izumi M, Nakamura A, Ikeda S, Takayama K, Yoshimura K, Nakagawa K, Yamamoto N, Sugio K. A Phase 2 Single-Arm Study of Osimertinib for Radiotherapy-Naive Central Nervous System Metastasis NSCLC: Results for the First-Line Cohort of the OCEAN Study (LOGIK 1603/WJOG 9116L). JTO Clin Res Rep 2023; 4:100587. [PMID: 38046380 PMCID: PMC10689268 DOI: 10.1016/j.jtocrr.2023.100587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/30/2023] [Accepted: 10/09/2023] [Indexed: 12/05/2023] Open
Abstract
Introduction Osimertinib may be effective in treating central nervous system (CNS) metastasis, but its efficacy in treating radiation therapy (RT)-naive metastasis is unclear. The OCEAN study assessed the efficacy of osimertinib against RT-naive CNS metastasis in patients previously treated (T790M cohort) and untreated patients (first-line cohort) with EGFR mutation. Here, we report the results of the first-line cohort. Methods Previously untreated patients with RT-naive CNS metastasis and EGFR mutation-positive NSCLC were treated with osimertinib. The brain metastasis response rate (BMRR), progression-free survival (PFS), and overall survival in the first-line cohort were secondary end points. Results A total of 26 patients were enrolled in the study between September 2019 and July 2020. The median age was 72.0 years with 80.8% female. There were 20 patients who had multiple CNS metastases. BMRR assessed by PAREXEL criteria was 76.9% (90% confidence interval [CI]: 63.3%-90.5%), BMRR assessed by Response Evaluation Criteria in Solid Tumors was 76.9% (95% CI: 54.0%-99.8%), and median PFS of CNS metastasis was 22.0 months (95% CI: 9.7 mo-not reached). The overall response rate was 64.0% (95% CI: 45.2%-82.8%), median PFS was 11.5 months (95% CI: 6.9 mo-not reached), and median survival time was 23.7 months (95% CI: 16.5 mo-not reached). Paronychia and increased creatinine level were the most frequent nonhematological toxicities observed in 13 patients (50%). Grade three and higher adverse events were less than 10%, and there were no treatment-related deaths. Pneumonitis was observed in five patients (19.2%). Conclusions These results suggest that osimertinib is effective in untreated patients with RT-naive asymptomatic CNS metastasis in a clinical practice first-line setting. Trial registration UMIN identifier: UMIN000024218. jRCT identifier: jRCTs071180017.
Collapse
Affiliation(s)
- Kazushige Wakuda
- Division of Thoracic Oncology, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Hiroyuki Yamaguchi
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hirotsugu Kenmotsu
- Division of Thoracic Oncology, Shizuoka Cancer Center Hospital, Shizuoka, Japan
| | - Minoru Fukuda
- Cancer Treatment Center, Nagasaki Prefecture Shimabara Hospital, Shimabara, Japan
| | - Kentaro Ito
- Department of Respiratory Medicine, Matsusaka Municipal Hospital Respiratory Center, Mie, Japan
| | - Yuko Tsuchiya-Kawano
- Department of Respiratory Medicine, Kitakyushu Municipal Medical Center, Fukuoka, Japan
| | - Kentaro Tanaka
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taishi Harada
- Department of Respiratory Medicine, Japan Community Health Care Organization (JCHO) Kyushu Hospital, Fukuoka, Japan
| | - Yuki Nakatani
- Department of Medical Oncology, Osaka City General Hospital, Osaka, Japan
| | - Satoru Miura
- Department of Internal Medicine, Niigata Cancer Center Hospital, Niigata, Japan
| | - Toshihide Yokoyama
- Department of Respiratory Medicine, Kurashiki Central Hospital, Okayama, Japan
| | - Tomomi Nakamura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Miiru Izumi
- Department of Respiratory Medicine, National Hospital Organization, Omuta National Hospital, Fukuoka, Japan
| | - Atsushi Nakamura
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Miyagi, Japan
| | - Satoshi Ikeda
- Department of Respiratory Medicine, Kanagawa Cardiovascular and Respiratory Center, Kanagawa, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenichi Yoshimura
- Center for Integrated Medical Research, Hiroshima University Hospital, Hiroshima University, Hiroshima, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | | | - Kenji Sugio
- Department of Thoracic and Breast Surgery, Oita University Faculty of Medicine, Oita, Japan
| |
Collapse
|
10
|
Iwama E, Yamamoto H, Okubo F, Ijichi K, Ibusuki R, Shiaraishi Y, Yoneshima Y, Tanaka K, Oda Y, Okamoto I. Evaluation of appropriate conditions for Oncomine DxTT testing of FFPE specimens for driver gene alterations in non-small cell lung cancer. Thorac Cancer 2023; 14:2288-2296. [PMID: 37345344 PMCID: PMC10423657 DOI: 10.1111/1759-7714.15014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND The Oncomine Dx Target Test Multi-CDx System (ODxTT) is a next-generation sequencing panel approved as a companion diagnostic for drugs targeted to corresponding gene alterations in non-small cell lung cancer. However, appropriate slide conditions for ODxTT have remained unclear. METHODS We focused on the production of the number of tumor cells on a formalin-fixed paraffin-embedded (FFPE) section and the number of prepared slides, designated the TS value, and determined a TS value of ≥4000 as a target slide condition for ODxTT. We evaluated the impact of this condition on ODxTT testing with tumor specimens found to have a TS of <4000 (n = 23) or a TS of ≥4000 (n = 142). RESULTS A positive correlation was apparent between the TS value and the concentrations of both DNA and RNA. Among the 142 samples with a TS of ≥4000, a sufficient concentration of DNA or RNA for ODxTT analysis was achieved in 100% and 98% samples, respectively. Among samples explored for driver gene alterations after determination of the target slide condition (TS ≥4000), most (84.9%) had a TS of ≥4000 and were submitted for ODxTT analysis. CONCLUSION Our findings indicate that a TS of ≥4000 is a feasible and relevant criterion for ODxTT testing, and its adoption should help to improve the success rate of such testing in clinical practice.
Collapse
Affiliation(s)
- Eiji Iwama
- Department of Respiratory Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Diagnostic PathologyKyushu University HospitalFukuokaJapan
- Department of PathologyOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| | - Fumihiko Okubo
- Division of Diagnostic PathologyKyushu University HospitalFukuokaJapan
| | - Kayo Ijichi
- Department of Respiratory Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Diagnostic PathologyKyushu University HospitalFukuokaJapan
| | - Ritsu Ibusuki
- Department of Respiratory Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yoshimasa Shiaraishi
- Department of Respiratory Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yasuto Yoneshima
- Department of Respiratory Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Kentaro Tanaka
- Department of Respiratory Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
- Division of Diagnostic PathologyKyushu University HospitalFukuokaJapan
| | - Isamu Okamoto
- Department of Respiratory Medicine, Graduate School of Medical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
11
|
Shen F, Guo W, Song X, Wang B. Molecular profiling and prognostic biomarkers in chinese non-small cell lung cancer cohort. Diagn Pathol 2023; 18:71. [PMID: 37301854 PMCID: PMC10257305 DOI: 10.1186/s13000-023-01349-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/25/2023] [Indexed: 06/12/2023] Open
Abstract
INTRODUCTION Comprehensive information about the genome analysis and its prognostic values of NSCLC patients in Chinese population are still needed. PATIENTS A total of 117 Chinese patients with NSCLC were enrolled in this study. Tumor tissues or blood were collected and sequenced by targeted next-generation sequencing of 556 cancer related genes. The associations between clinical outcomes and clinical characteristics, TMB, mutated genes, treatment therapies were analyzed using Kaplan-Meier methods and further evaluated using multivariable Cox proportional hazards regression model. RESULTS A total of 899 mutations were identified by targeted NGS. The most frequently mutations included EGFR (47%), TP53 (46%), KRAS (18%), LRP1B (12%) and SPTA1 (10%). Patients with mutant TP53, PREX2, ARID1A, PTPRT and PIK3CG had lower median overall survival (OS) than those patients with wild-type (P = 0.0056, P < 0.001, P < 0.0001, P < 0.0001 and P = 0.036, respectively). Using a multivariate Cox regression model, PREX2 (P < 0.001), ARID1A (P < 0.001) and PIK3CG (P = 0.04) were independent prognostic factors in NSCLC. In the patients received chemotherapy, squamous patients had a significantly longer median OS than adenocarcinoma patients (P = 0.011). In the patients received targeted therapy, adenocarcinoma patients had a significantly longer survival period than squamous patients (P = 0.01). CONCLUSIONS Our study provided comprehensive genomic alterations in a cohort of Chinese NSCLC. We also identified new prognostic biomarkers, which could provide potential clues for targeted therapies.
Collapse
Affiliation(s)
- Fangfang Shen
- Department of Respiratory Medicine, Shanxi Hospital Affiliated to Cancer Hospital, Affiliated Cancer Hospital of Shanxi Medical University, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, 030001, China
| | - Wei Guo
- Department of Respiratory Medicine, Shanxi Hospital Affiliated to Cancer Hospital, Affiliated Cancer Hospital of Shanxi Medical University, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, 030001, China
| | - Xia Song
- Department of Respiratory Medicine, Shanxi Hospital Affiliated to Cancer Hospital, Affiliated Cancer Hospital of Shanxi Medical University, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, 030001, China
| | - Bei Wang
- The Second Hospital, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
12
|
Le X, Elamin YY, Zhang J. New Actions on Actionable Mutations in Lung Cancers. Cancers (Basel) 2023; 15:cancers15112917. [PMID: 37296880 DOI: 10.3390/cancers15112917] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 05/24/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Actionable mutations refer to DNA alterations that, if detected, would be expected to affect patients' response to treatments [...].
Collapse
Affiliation(s)
- Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yasir Y Elamin
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
13
|
Poveda-Rogers C, Morrissette JJD. Greater expectations: meeting clinical needs through broad and rapid genomic testing. Clin Chem Lab Med 2023; 61:654-661. [PMID: 36473133 DOI: 10.1515/cclm-2022-1016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
Cancer describes a group of diseases driven by genetic and genomic changes that can occur across hundreds of different genes. Knowledge of the specific variants present in a patient's cancer can help to predict response to different treatment options, confirm disease diagnosis, and understand a patient's prognosis and risks, which ultimately leads to improved survival outcomes. The advent of next-generation sequencing (NGS) technology has allowed pathologists to simultaneously profile the sequences of many genes in a single reaction, but not all NGS assays are built the same. While those used for broad genomic profiling are useful to probe large regions of the genome and gather more information about a patient's tumor, it comes at the cost of relatively long turnaround times (TAT), which may be detrimental to patient care. Conversely, NGS assays used for rapid genomic profiling provide faster results, but may miss detection of variants that are clinically informative. Determining which type of genomic profiling to order depends on a number of factors including the severity of a patient's illness, standard of care paradigms, and success or failure of previous therapies. Ultimately, the ideal clinical diagnostic laboratory will be able to offer both options to best meet the clinical needs of its patients.
Collapse
Affiliation(s)
- Corey Poveda-Rogers
- Hospital of the University of Pennsylvania, Pathology and Laboratory Medicine, Philadelphia, PA, USA
| | - Jennifer J D Morrissette
- Hospital of the University of Pennsylvania, Pathology and Laboratory Medicine, Philadelphia, PA, USA
| |
Collapse
|
14
|
Zhang C, Kim RY, McGrath CM, Andronov M, Haas AR, Ma KC, Lanfranco AR, Hutchinson CT, Morrissette JJD, DiBardino DM. The Performance of an Extended Next Generation Sequencing Panel Using Endobronchial Ultrasound-Guided Fine Needle Aspiration Samples in Non-Squamous Non-Small Cell Lung Cancer: A Pragmatic Study. Clin Lung Cancer 2023; 24:e105-e112. [PMID: 36599742 PMCID: PMC10664188 DOI: 10.1016/j.cllc.2022.11.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022]
Abstract
INTRODUCTION/BACKGROUND Samples from endobronchial ultrasound-guided fine needle aspiration (EBUS-TBNA) are frequently used for next generation sequencing (NGS) in patients with non-small cell lung cancer (NSCLC) to look for genetic driver mutations. The objective of the current study was to evaluate the performance of extended NGS panels using EBUS-TBNA samples in a real-world setting and identify factors associated with the success of NGS. MATERIALS AND METHODS This study included all patients who underwent EBUS and were diagnosed with non-squamous NSCLC with mediastinal metastasis from 2016 to 2019 at the University of Pennsylvania. We reviewed demographic information, imaging studies, procedure reports, pathology and NGS reports. Logistic regression was used to analyze factors associated with the success of NGS panels. RESULTS The success rates of NGS using EBUS-TBNA samples were 92.5%, and 91.5% for DNA and RNA NGS panels respectively. Samples from higher N stage (N2 and N3 lymph nodes) and with higher tumor cellularity (>25%) resulted in higher success rate for DNA NGS. The effect of tumor cellularity remained borderline significant after entering multivariable logistic regression. The short-axis diameter of the sampled lymph node on CT scan, FDG-avidity on PET CT and >3 EBUS passes per lymph node during the procedure were not associated with NGS success. CONCLUSION Both DNA and RNA extended-panel NGS had high performance using EBUS-TBNA samples. Sampling more advanced nodal stations and obtaining samples with higher tumor cellularity were associated with higher success rate of DNA NGS. Other imaging or procedural factors did not affect NGS performance.
Collapse
Affiliation(s)
- Chenchen Zhang
- Division of Thoracic Surgery & Interventional Pulmonology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA.
| | - Roger Y Kim
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Cindy M McGrath
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Michelle Andronov
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Andrew R Haas
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Kevin C Ma
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Anthony R Lanfranco
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Christoph T Hutchinson
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Jennifer J D Morrissette
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - David M DiBardino
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
15
|
Raez LE, Brice K, Dumais K, Lopez-Cohen A, Wietecha D, Izquierdo PA, Santos ES, Powery HW. Liquid Biopsy Versus Tissue Biopsy to Determine Front Line Therapy in Metastatic Non-Small Cell Lung Cancer (NSCLC). Clin Lung Cancer 2023; 24:120-129. [PMID: 36585341 DOI: 10.1016/j.cllc.2022.11.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 11/27/2022]
Abstract
In the last decade, non-small-cell lung cancer (NSCLC) treatment has improved with the approval of multiple therapies to target specific genetic alterations. Though, next generation sequencing (NGS) has traditionally been conducted from tissue biopsy samples, developing data supports the use of plasma-based circulating tumor DNA (ctDNA), also known as "liquid biopsy," to complement tissue biopsy approaches in guiding front-line therapy. This study is a retrospective analysis of 170 new NSCLC patients treated at 2 cancer centers within a 5-year period who received both tissue and liquid biopsy NGS as standard of care. Based on a treatment schema defined by testing sufficiency, biomarker detection, and turnaround time (TAT), physicians based the majority of their treatments on liquid biopsy results (73.5%) versus tissue biopsy (25.9%). Liquid biopsy NGS returned results on average 26.8 days faster than tissue and reported higher testing success. For guideline-recommended biomarkers, liquid biopsy was 94.8% to 100% concordant with tissue. In comparing testing modalities, a liquid-first approach identified guideline-recommended biomarkers in 76.5% of patients versus 54.9% in a tissue-first approach. There was no significant difference in time-to-treatment, or survival outcomes (overall survival and progression free survival) based on liquid versus tissue biopsy findings. This research demonstrates that liquid biopsy NGS is an effective tool to capture actionable genetic alterations in NSCLC. Due to its high concordance to tissue, faster TAT, and similarity in outcomes and time-to-treatment, liquid biopsy can be used either as a first-line test or concordantly with tissue biopsy to guide treatment decisions in NSCLC.
Collapse
Affiliation(s)
- Luis E Raez
- Thoracic Oncology Program, Memorial Cancer Institute/Florida Atlantic University, Pembroke Pines, FL.
| | - Kayla Brice
- Memorial Cancer Institute, Pembroke Pines, FL
| | | | | | - Delia Wietecha
- Department of Medicine/Memorial Healthcare System, Pembroke Pines, FL
| | | | - Edgardo S Santos
- Florida Precision Oncology/Genesis Care/Florida Atlantic University
| | | |
Collapse
|
16
|
Simons MJHG, Uyl-de Groot CA, Retèl VP, Mankor JM, Ramaekers BLT, Joore MA, van Harten WH. Cost-Effectiveness and Budget Impact of Future Developments With Whole-Genome Sequencing for Patients With Lung Cancer. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2023; 26:71-80. [PMID: 35973926 DOI: 10.1016/j.jval.2022.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 06/19/2022] [Accepted: 07/08/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVES This study aimed to investigate the cost-effectiveness, budget impact (BI), and impact of uncertainty of future developments concerning whole-genome sequencing (WGS) as a clinical diagnostic test compared with standard of care (SoC) in patients with locally advanced and metastatic non-small cell lung cancer. METHODS A total of 3 likely scenarios to take place within 5 years (according to experts) were simulated using a previously developed, peer reviewed, and published decision model. The scenarios concerned "WGS results used for treatment selection" (scenario 1), "WGS-based biomarker for immunotherapy" (scenario 2), and "off-label drug approval for WGS results" (scenario 3). Two diagnostic strategies of the original model, "SoC" and "WGS as a diagnostic test" (base model), were used to compare our scenarios with. Outcomes were reported for the base model, all scenarios separately, combined (combined unweighted), and weighted by likelihood (combined weighted). Cost-effectiveness, BI, and value of information analyses were performed for WGS compared with SoC. RESULTS Total costs and quality-adjusted life-years for SoC in metastatic non-small cell lung cancer were €149 698 and 1.235. Incremental outcomes of WGS were €1529/0.002(base model), -€222/0.020(scenario 1), -€2576/0.023(scenario 2), €388/0.024(scenario 3), -€5041/0.060(combined unweighted), and -€1715/0.029(combined weighted). The annual BI for adopting WGS for this population in The Netherlands ranged between €682 million (combined unweighted) and €714 million (base model). The consequences of uncertainty amounted to €3.4 million for all scenarios (combined weighted) and to €699 000 for the diagnostic yield of WGS alone (combined weighted). CONCLUSIONS Our findings suggest that it is likely for WGS to become cost-effective within the near future if it identifies more patients with actionable targets and show the impact of uncertainty regarding its diagnostic yield. Modeling future scenarios can be useful to consider early adoption of WGS while timely anticipating on unforeseen developments before final conclusions are reached.
Collapse
Affiliation(s)
- Martijn J H G Simons
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre, Maastricht, The Netherlands; Care And Public Health Research Institute, Maastricht University, Maastricht, The Netherlands
| | - Carin A Uyl-de Groot
- Erasmus School of Health Policy and Management/Institute for Medical Technology Assessment, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Valesca P Retèl
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek hospital, Amsterdam, The Netherlands; Department of Health Technology and Services Research, University of Twente, Enschede, The Netherlands
| | - Joanne M Mankor
- Department of Pulmonary Medicine, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Bram L T Ramaekers
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre, Maastricht, The Netherlands; Care And Public Health Research Institute, Maastricht University, Maastricht, The Netherlands
| | - Manuela A Joore
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre, Maastricht, The Netherlands; Care And Public Health Research Institute, Maastricht University, Maastricht, The Netherlands.
| | - Wim H van Harten
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek hospital, Amsterdam, The Netherlands; Department of Health Technology and Services Research, University of Twente, Enschede, The Netherlands
| |
Collapse
|
17
|
Buglioni A, Caffes PL, Hessler MG, Mansfield AS, Lo YC. Clinical Utility Validation of an Automated Ultrarapid Gene Fusion Assay for NSCLC. JTO Clin Res Rep 2022; 3:100434. [PMID: 36536899 PMCID: PMC9758522 DOI: 10.1016/j.jtocrr.2022.100434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/13/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
Abstract
Introduction Gene rearrangements are frequent oncologic drivers in NSCLC, and many are suitable for treatment with Food and Drug Administration-approved or experimental targeted therapies. We evaluated the accuracy, specimen acceptance profile, and limits of detection of a rapid fusion assay (Idylla GeneFusion Assay), a commercially available ultrarapid molecular assay, for its clinical utility. Methods A collection of 97 specimens which had previously undergone next-generation sequencing testing were analyzed using the rapid fusion assay. Accuracy was evaluated by sensitivity and specificity compared with the next-generation sequencing results. The performance characteristics were tested by using a variety of different clinically relevant specimen types. Limits of detection were assessed by evaluating different input of tumor percentage and material amount. Results The rapid fusion assay was found to have 100% sensitivity in detecting fusions of ALK, ROS1, RET, NTRK1, and MET exon 14 skipping and 83% sensitivity for NTRK2/3 fusions. There were 100% specificity in detecting fusions of ROS1, RET, NTRK2/3, and MET exon 14 skipping and 98% specificity for ALK. Testing was successful with formalin-fixed paraffin-embedded biopsy and surgical tissues, cell blocks from fine-needle aspiration and pleural fluid (down to 5% tumor content, 18 mm2 tissue scraped), cytology smears (≥300 cells), and previously extracted RNA (minimal 20 ng). Conclusions The rapid fusion assay is quick, accurate, and versatile, allowing reliable detection of ALK, ROS1, RET fusions, and MET exon 14 skipping in NSCLC, and NTRK fusions. Rapid molecular testing may expedite treatment with appropriate targeted therapies.
Collapse
Affiliation(s)
- Alessia Buglioni
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Patricia L. Caffes
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Mark G. Hessler
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | - Ying-Chun Lo
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
18
|
Cascetta P, Marinello A, Lazzari C, Gregorc V, Planchard D, Bianco R, Normanno N, Morabito A. KRAS in NSCLC: State of the Art and Future Perspectives. Cancers (Basel) 2022; 14:5430. [PMID: 36358848 PMCID: PMC9656434 DOI: 10.3390/cancers14215430] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 07/30/2023] Open
Abstract
In NSCLC, KRAS mutations occur in up to 30% of all cases, most frequently at codon 12 and 13. KRAS mutations have been linked to adenocarcinoma histology, positive smoking history, and Caucasian ethnicity, although differences have been described across KRAS mutational variants subtypes. KRAS mutations often concur with other molecular alterations, notably TP53, STK11, and KEAP1, which could play an important role in treatment efficacy and patient outcomes. For many years, KRAS mutations have been considered undruggable mainly due to a high toxicity profile and low specificity of compounds. Sotorasib and adagrasib are novel KRAS inhibitors that recently gained FDA approval for pre-treated KRAS mutant NSCLC patients, and other molecules such as GDC-6036 are currently being investigated with promising results. Despite their approval, the efficacy of these drugs is lower than expected and progression among responders has been reported. Mechanisms of acquired resistance to anti-KRAS molecules typically involves either on target secondary mutations (e.g., G12, G13, Q61H, R68S, H95, Y96C, V8L) or off-target alterations. Ongoing trials are currently evaluating strategies for implementing efficacy and overcoming acquired resistance to these compounds. Finally, the efficacy of immune-checkpoint inhibitors still needs to be completely assessed and responses to anti-PD-1/PD-L1 agents may strongly depend on concomitant mutations.
Collapse
Affiliation(s)
- Priscilla Cascetta
- Department of Medical Oncology, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94850 Villejuif, France
| | - Arianna Marinello
- Department of Medical Oncology, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94850 Villejuif, France
- Department of Medical Oncology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Italy
| | - Chiara Lazzari
- Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy
| | - Vanesa Gregorc
- Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy
| | - David Planchard
- Department of Medical Oncology, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, 94850 Villejuif, France
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, Oncology Division, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Nicola Normanno
- Cellular Biology and Biotherapy, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Via Mariano Semmola 53, 80131 Naples, Italy
| | - Alessandro Morabito
- Thoracic Medical Oncology, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Via Mariano Semmola 53, 80131 Naples, Italy
| |
Collapse
|
19
|
Tabti K, Elmchichi L, Sbai A, Maghat H, Bouachrine M, Lakhlifi T. Molecular modelling of antiproliferative inhibitors based on SMILES descriptors using Monte-Carlo method, docking, MD simulations and ADME/Tox studies. MOLECULAR SIMULATION 2022. [DOI: 10.1080/08927022.2022.2110246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Affiliation(s)
- Kamal Tabti
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| | - Larbi Elmchichi
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| | - Abdelouahid Sbai
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| | - Hamid Maghat
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| | - Mohammed Bouachrine
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, Moulay Ismail University, Meknes, Morocco
- High School of Technology Khenifra, Sultan Moulay Sliman University, Benimellal, Morocco
| | - Tahar Lakhlifi
- Molecular Chemistry and Natural Substances Laboratory, Faculty of Science, Moulay Ismail University, Meknes, Morocco
| |
Collapse
|
20
|
Feng A, Li Y, Li G, Wang Y, Wen Q, Yang Z, Tian K, Lv H, Guo L, Zhang S, Liu X, Jiang D. Genomic Features of Organ-Specific Metastases in Lung Adenocarcinoma. Front Oncol 2022; 12:908759. [PMID: 35912232 PMCID: PMC9331737 DOI: 10.3389/fonc.2022.908759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/09/2022] [Indexed: 12/24/2022] Open
Abstract
BackgroundThe genomic features of cancer cells may confer the metastatic ability of lung adenocarcinoma (LUAD) to metastasize to specific organs. We aimed to identify the differences in genomic alterations between patients with primary LUAD with and without metastases and to elucidate the metastatic biology that may help developing biomarker-directed therapies for advanced or metastatic disease.MethodsA retrospective cohort of 497 patients with LUAD including 388 primary tumors (PR), 53 bone metastases (MT-bone), 30 liver metastases (MT-liver), and 26 brain metastases (MT-brain) was tested for genomic alterations by a next-generation sequencing assay.ResultsThe EGFR, TP53, TERT, LRP1B, CDKN2A, ERBB2, ALK, and KMT2C genes had a high frequency of mutations, and the mutations were shared by PR and metastases groups. TP53 and EGFR were the most common mutated genes. In comparison with PR, KRAS, STK11, ATM, NPM1, and ROS1 were significantly mutated in MT-brain, and TP53, MYC, RSPO2, CDKN2a, and CDKN2B were significantly mutated in MT-liver. The frequencies of TP53, CDKN2A, MTAP, PRKCI, and APC mutations were higher in MT-bone than that in PR. The ERBB, phosphoinositide-3-kinase/protein kinase B (PI3K-AKT), cell cycle, Fibroblast growth factor (FGF), and homologous recombination deficiency signaling pathways were affected in both PR and metastases, and there is higher frequency of mutations in metastases. Moreover, the co-mutations in patients with PR and metastasis were respectively analyzed. In addition, the programmed death ligand 1 (PD-L1) level was obviously related to tumor stage and tumor metastases, and the tumor mutational burden was correlated to clinicopathological features including age, gender, pathological stages, and tumor metastases. FGFR1, KAT6A, MYC, RAD21, TP53, and DAXX were also dramatically correlated to the tumor mutational burden.ConclusionMetastases are the most devastating stage of tumors and the main cause of cancer-related deaths. Our results provided a clinically relevant view of the tumor-intrinsic mutational landscape of patients with metastatic LUAD.
Collapse
Affiliation(s)
- Alei Feng
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Qidu Pharmaceutical Co. Ltd., Shandong Provincial Key Laboratory of Neuroprotective Drugs, Zibo, China
| | - Yanjun Li
- Department of Thoracic Surgery, The Second People’s Hospital of Dezhou, Dezhou, China
| | - Guangxu Li
- Department of Thoracic Surgery, The Second People’s Hospital of Dezhou, Dezhou, China
| | - Yu Wang
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qiang Wen
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhe Yang
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Kaihua Tian
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongying Lv
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lijie Guo
- Shanghai OrigiMed Co., Ltd, Shanghai, China
| | | | | | - Da Jiang
- Medical Oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Da Jiang,
| |
Collapse
|
21
|
Zhao X, Cui L, Zhang Y, Guo C, Deng L, Wen Z, Lu Z, Shi X, Xing H, Liu Y, Zhang Y. Screening for Potential Therapeutic Agents for Non-Small Cell Lung Cancer by Targeting Ferroptosis. Front Mol Biosci 2022; 9:917602. [PMID: 36203872 PMCID: PMC9532010 DOI: 10.3389/fmolb.2022.917602] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Ferroptosis is a form of non-apoptotic and iron-dependent cell death originally identified in cancer cells. Recently, emerging evidence showed that ferroptosis-targeting therapy could be a novel promising anti-tumour treatment. However, systematic analyses of ferroptosis-related genes for the prognosis of non-small cell lung cancer (NSCLC) and the development of antitumor drugs exploiting the ferroptosis process remain rare. This study aimed to identify genes related to ferroptosis and NSCLC and to initially screen lead compounds that induce ferroptosis in tumor cells. We downloaded mRNA expression profiles and NSCLC clinical data from The Cancer Genome Atlas database to explore the prognostic role of ferroptosis-related genes. Four prognosis-associated ferroptosis-related genes were screened using univariate Cox regression analysis and the lasso Cox regression analysis, which could divide patients with NSCLC into high- and low-risk groups. Then, based on differentially expressed risk- and ferroptosis-related genes, the negatively correlated lead compound flufenamic acid (FFA) was screened through the Connective Map database. This project confirmed that FFA induced ferroptosis in A549 cells and inhibited growth and migration in a dose-dependent manner through CCK-8, scratch, and immunofluorescence assays. In conclusion, targeting ferroptosis might be a therapeutic alternative for NSCLC.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Lijuan Cui
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Yushan Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Chao Guo
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Lijiao Deng
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Zhitong Wen
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Zhihong Lu
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
- Department of Endocrinology, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| | - Xiaoyuan Shi
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Haojie Xing
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Yunfeng Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
- *Correspondence: Yunfeng Liu, ; Yi Zhang, , orcid.org/0000-0003-0305-3127
| | - Yi Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
- *Correspondence: Yunfeng Liu, ; Yi Zhang, , orcid.org/0000-0003-0305-3127
| |
Collapse
|
22
|
Pedrini E, Negro A, Di Brino E, Pecoraro V, Sculco C, Abelli E, Gnoli M, Magrelli A, Sangiorgi L, Cicchetti A. Real-World Data and Budget Impact Analysis (BIA): Evaluation of a Targeted Next-Generation Sequencing Diagnostic Approach in Two Orthopedic Rare Diseases. Front Pharmacol 2022; 13:785705. [PMID: 35734413 PMCID: PMC9207266 DOI: 10.3389/fphar.2022.785705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Next-generation sequencing (NGS) technology, changing the diagnostic approach, has become essential in clinical settings, and its adoption by public health laboratories is now the practice. Despite this, as technological innovations, its intake requires an evaluation of both the clinical utility and the economic investment, especially considering the rare disease scenario. This study evaluated the analytical validity and the budget impact of an NGS-Ion Torrent™ approach for the molecular germline diagnosis of two musculoskeletal rare diseases.Methods: Two cohorts of 200 and 199 patients with suspect or clinical diagnosis of multiple osteochondromas (MO) and osteogenesis imperfecta (OI) previously evaluated with a single-gene diagnostic protocol were re-analyzed using a targeted NGS assay. Analytical validity was assessed by comparing NGS and single-gene protocol. A budget impact analysis using real-world cost data-considering the healthcare perspective— was performed by applying activity-based costing (ABC). The cost considered consumables, personnel, and equipment. Additional costs not related to NGS activities were not considered. Sensitivity analysis was performed.Results: The NGS method showed a higher (for MO) and comparable (for OI) diagnostic sensitivity than the traditional techniques, apart from always reducing the time and costs of diagnosis. Overall, the cost saving per patient is € 765 for OI and € 74 for MO. Materials represented the highest cost driver of the NGS process. A time saving—proportional to the panel size—has been assessed in both cases.Conclusions: Our targeted NGS diagnostic approach decreases time to diagnosis and costs, appearing to be beneficial and recommended both for patients and from a healthcare perspective in routine diagnosis also considering very small gene panels and a low patient flow. The adequate analytical sensitivity always required the additional Sanger sequencing step of the low- and non-covered regions. A more accurate strategy evaluation is suggested in the case of ultra-rare/complex diseases, large gene-panel, or non-reference diagnostic centers.
Collapse
Affiliation(s)
- Elena Pedrini
- Department of Rare Skeletal Disorders, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Antonella Negro
- Regional Agency for Health and Social Care of Emilia-Romagna, Bologna, Italy
| | - Eugenio Di Brino
- Università Cattolica del Sacro Cuore, Graduate School of Health Economics and Management, Rome, Italy
| | - Valentina Pecoraro
- Regional Agency for Health and Social Care of Emilia-Romagna, Bologna, Italy
| | - Camilla Sculco
- School of Economics and Management, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Elisabetta Abelli
- Department of Rare Skeletal Disorders, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Maria Gnoli
- Department of Rare Skeletal Disorders, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Armando Magrelli
- National Center for Drug Research and Evaluation, National Institute of Health (ISS), Rome, Italy
| | - Luca Sangiorgi
- Department of Rare Skeletal Disorders, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- *Correspondence: Luca Sangiorgi,
| | - Americo Cicchetti
- Università Cattolica del Sacro Cuore, Graduate School of Health Economics and Management, Rome, Italy
| |
Collapse
|
23
|
Ryan DJ, Toomey S, Smyth R, Madden SF, Workman J, Cummins R, Sheehan K, Fay J, Naidoo J, Breathnach OS, Morris PG, Grogan L, O'Brien ME, Sulaiman I, Hennessy BT, Morgan RK. Exhaled Breath Condensate (EBC) analysis of circulating tumour DNA (ctDNA) using a lung cancer specific UltraSEEK oncogene panel. Lung Cancer 2022; 168:67-73. [DOI: 10.1016/j.lungcan.2022.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 10/18/2022]
|
24
|
Kang S, Woo J, Kim S. A Systematic Review of Companion Diagnostic Tests by Immunohistochemistry for the Screening of Alectinib-Treated Patients in ALK-Positive Non-Small Cell Lung Cancer. Diagnostics (Basel) 2022; 12:diagnostics12051297. [PMID: 35626451 PMCID: PMC9140374 DOI: 10.3390/diagnostics12051297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/13/2022] [Accepted: 05/20/2022] [Indexed: 11/29/2022] Open
Abstract
Companion diagnostic tests and targeted therapy changed the management of non-small cell lung cancer by diagnosing genetic modifications and enabling individualized treatment. The purpose of this systematic review is to assess the clinical applicability of companion diagnostic tests (IHC method) by comparing the effects of alectinib and crizotinib in patients with ALK-positive NSCLC. We searched for literature up to March 2022 in PubMed, Web of Science, Cochrane, and Google Scholar. The inclusion criteria were randomized controlled trials comparing the effectiveness of alectinib and crizotinib using an IHC-based companion diagnostic test. The primary outcome was progression-free survival (PFS). The secondary outcomes were objective response rate (ORR), duration of response (DOR), and overall survival (OS). PFS was longer in alectinib (68.4 [61.0, 75.9]) than crizotinib (48.7 [40.4, 56.9]). This indicated that alectinib had a superior efficacy to that of crizotinib (HR range 0.15–0.47). In all secondary outcomes, alectinib was better than crizotinib. Particularly for the ORR, the odds ratio (OR) confirmed that alectinib had a lower risk rate (OR: 2.21, [1.46–3.36], p = 0.0002, I2 = 39%). Therefore, the companion diagnostic test (immunohistochemistry) is an effective test to determine whether to administer alectinib to ALK-positive NSCLC patients.
Collapse
Affiliation(s)
- Sulim Kang
- Department of Medical Industry, Dongguk University-Seoul, 26, Pil-dong 3-ga, Jung-gu, Seoul 04620, Korea; (S.K.); (J.W.)
| | - Jaehyun Woo
- Department of Medical Industry, Dongguk University-Seoul, 26, Pil-dong 3-ga, Jung-gu, Seoul 04620, Korea; (S.K.); (J.W.)
| | - Sungmin Kim
- Department of Medical Industry, Dongguk University-Seoul, 26, Pil-dong 3-ga, Jung-gu, Seoul 04620, Korea; (S.K.); (J.W.)
- Department of Medical Biotechnology, Dongguk University, Bio Medi Campus, 32, Dongguk-ro, Ilsandong-gu, Goyang-si 10326, Gyeonggi-do, Korea
- Correspondence:
| |
Collapse
|
25
|
Poh J, Ngeow KC, Pek M, Tan KH, Lim JS, Chen H, Ong CK, Lim JQ, Lim ST, Lim CM, Goh BC, Choudhury Y. Analytical and clinical validation of an amplicon-based next generation sequencing assay for ultrasensitive detection of circulating tumor DNA. PLoS One 2022; 17:e0267389. [PMID: 35486650 PMCID: PMC9053827 DOI: 10.1371/journal.pone.0267389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 04/07/2022] [Indexed: 11/19/2022] Open
Abstract
Next-generation sequencing of circulating tumor DNA presents a promising approach to cancer diagnostics, complementing conventional tissue-based diagnostic testing by enabling minimally invasive serial testing and broad genomic coverage through a simple blood draw to maximize therapeutic benefit to patients. LiquidHALLMARK® is an amplicon-based next-generation sequencing assay developed for the genomic profiling of plasma-derived cell-free DNA (cfDNA). The comprehensive 80-gene panel profiles point mutations, insertions/deletions, copy number alterations, and gene fusions, and further detects oncogenic viruses (Epstein-Barr virus (EBV) and hepatitis B virus (HBV)) and microsatellite instability (MSI). Here, the analytical and clinical validation of the assay is reported. Analytical validation using reference genetic materials demonstrated a sensitivity of 99.38% for point mutations and 95.83% for insertions/deletions at 0.1% variant allele frequency (VAF), and a sensitivity of 91.67% for gene fusions at 0.5% VAF. In non-cancer samples, a high specificity (≥99.9999% per-base) was observed. The limit of detection for copy number alterations, EBV, HBV, and MSI were also empirically determined. Orthogonal comparison of epidermal growth factor receptor (EGFR) variant calls made by LiquidHALLMARK and a reference allele-specific polymerase chain reaction (AS-PCR) method for 355 lung cancer specimens revealed an overall concordance of 93.80%, while external validation with cobas® EGFR Mutation Test v2 for 50 lung cancer specimens demonstrated an overall concordance of 84.00%, with a 100% concordance rate for EGFR variants above 0.4% VAF. Clinical application of LiquidHALLMARK in 1,592 consecutive patients demonstrated a high detection rate (74.8% circulating tumor DNA (ctDNA)-positive in cancer samples) and broad actionability (50.0% of cancer samples harboring alterations with biological evidence for actionability). Among ctDNA-positive lung cancers, 72.5% harbored at least one biomarker with a guideline-approved drug indication. These results establish the high sensitivity, specificity, accuracy, and precision of the LiquidHALLMARK assay and supports its clinical application for blood-based genomic testing.
Collapse
Affiliation(s)
- Jonathan Poh
- Lucence Diagnostics Pte Ltd, Singapore, Singapore
| | | | - Michelle Pek
- Lucence Health Inc, Palo Alto, California, United States of America
| | - Kian-Hin Tan
- Lucence Health Inc, Palo Alto, California, United States of America
| | | | - Hao Chen
- Lucence Diagnostics Pte Ltd, Singapore, Singapore
| | | | - Jing Quan Lim
- National Cancer Centre Singapore, Singapore, Singapore
| | - Soon Thye Lim
- National Cancer Centre Singapore, Singapore, Singapore
| | - Chwee Ming Lim
- Department of Otolaryngology-Head and Neck Surgery, Singapore General Hospital, Duke-NUS Medical School, Singapore, Singapore
| | | | | |
Collapse
|
26
|
Karadzovska-Kotevska M, Brunnström H, Kosieradzki J, Ek L, Estberg C, Staaf J, Barath S, Planck M. Feasibility of EBUS-TBNA for histopathological and molecular diagnostics of NSCLC-A retrospective single-center experience. PLoS One 2022; 17:e0263342. [PMID: 35108331 PMCID: PMC8809531 DOI: 10.1371/journal.pone.0263342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/17/2022] [Indexed: 12/24/2022] Open
Abstract
Endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) is a minimally invasive bronchoscopic procedure, well established as a diagnostic modality of first choice for diagnosis and staging of non-small cell lung cancer (NSCLC). The therapeutic decisions for advanced NSCLC require comprehensive profiling of actionable mutations, which is currently considered to be an essential part of the diagnostic process. The purpose of this study was to evaluate the utility of EBUS-TBNA cytology specimen for histological subtyping, molecular profiling of NSCLC by massive parallel sequencing (MPS), as well as for PD-L1 analysis. A retrospective review of 806 EBUS bronchoscopies was performed, resulting in a cohort of 132 consecutive patients with EBUS-TBNA specimens showing NSCLC cells in lymph nodes. Data on patient demographics, radiology features of the suspected tumor and mediastinal engagement, lymph nodes sampled, the histopathological subtype of NSCLC, and performed molecular analysis were collected. The EBUS-TBNA specimen proved sufficient for subtyping NSCLC in 83% and analysis of treatment predictive biomarkers in 77% (MPS in 53%). The adequacy of the EBUS-TBNA specimen was 69% for EGFR gene mutation analysis, 49% for analysis of ALK rearrangement, 36% for ROS1 rearrangement, and 33% for analysis of PD-L1. The findings of our study confirm that EBUS-TBNA cytology aspirate is appropriate for diagnosis and subtyping of NSCLC and largely also for treatment predictive molecular testing, although more data is needed on the utility of EBUS cytology specimen for MPS and PD-L1 analysis.
Collapse
Affiliation(s)
- Marija Karadzovska-Kotevska
- Department of Respiratory Diseases and Allergology, Skåne University Hospital Lund, Lund, Sweden
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Lund, Sweden
- * E-mail:
| | - Hans Brunnström
- Division of Laboratory Medicine, Department of Genetics and Pathology, Region Skåne, Lund, Sweden
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Jaroslaw Kosieradzki
- Department of Respiratory Diseases and Allergology, Skåne University Hospital Lund, Lund, Sweden
| | - Lars Ek
- Department of Respiratory Diseases and Allergology, Skåne University Hospital Lund, Lund, Sweden
| | - Christel Estberg
- Department of Respiratory Diseases and Allergology, Skåne University Hospital Lund, Lund, Sweden
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Lund, Sweden
| | - Stefan Barath
- Department of Respiratory Diseases and Allergology, Skåne University Hospital Lund, Lund, Sweden
| | - Maria Planck
- Department of Respiratory Diseases and Allergology, Skåne University Hospital Lund, Lund, Sweden
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, Lund, Sweden
| |
Collapse
|
27
|
Simons MJHG, Retèl VP, Ramaekers BLT, Butter R, Mankor JM, Paats MS, Aerts JGJV, Mfumbilwa ZA, Roepman P, Coupé VMH, Uyl-de Groot CA, van Harten WH, Joore MA. Early Cost Effectiveness of Whole-Genome Sequencing as a Clinical Diagnostic Test for Patients with Inoperable Stage IIIB,C/IV Non-squamous Non-small-Cell Lung Cancer. PHARMACOECONOMICS 2021; 39:1429-1442. [PMID: 34405371 PMCID: PMC8599348 DOI: 10.1007/s40273-021-01073-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/25/2021] [Indexed: 05/09/2023]
Abstract
BACKGROUND Advanced non-small-cell lung cancer (NSCLC) harbours many genetic aberrations that can be targeted with systemic treatments. Whole-genome sequencing (WGS) can simultaneously detect these (and possibly new) molecular targets. However, the exact added clinical value of WGS is unknown. OBJECTIVE The objective of this study was to determine the early cost effectiveness of using WGS in diagnostic strategies compared with currently used molecular diagnostics for patients with inoperable stage IIIB,C/IV non-squamous NSCLC from a Dutch healthcare perspective. METHODS A decision tree represented the diagnostic pathway, and a cohort state transition model represented disease progression. Three diagnostic strategies were modelled: standard of care (SoC) alone, WGS as a diagnostic test, and SoC followed by WGS. Treatment effectiveness was based on a systematic review. Probabilistic cost-effectiveness analyses were performed, and threshold analyses (using €80,000 per quality-adjusted life-year [QALY]) was used to explore the early cost effectiveness of WGS. RESULTS WGS as a diagnostic test resulted in more QALYs (0.002) and costs (€1534 [incremental net monetary benefit -€1349]), and SoC followed by WGS resulted in fewer QALYs (-0.002) and more costs (€1059 [-€1194]) compared with SoC alone. WGS as a diagnostic test was only cost effective if it was priced at €2000 per patient and identified 2.7% more actionable patients than SoC alone. Treating these additional identified patients with new treatments costing >€4069 per month decreased the probability of cost effectiveness. CONCLUSIONS Our analysis suggests that providing WGS as a diagnostic test is cost effective compared with SoC followed by WGS and SoC alone if costs for WGS decrease and additional patients with actionable targets are identified. This cost-effectiveness model can be used to incorporate new findings iteratively and to support ongoing decision making regarding the use of WGS in this rapidly evolving field.
Collapse
Affiliation(s)
- Martijn J H G Simons
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre, P. Debyelaan 25, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
- Maastricht University, Care and Public Health Research Institute (CAPHRI), Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands
| | - Valesca P Retèl
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Health Technology and Services Research, University of Twente, Hallenweg 5, 7522 NH, Enschede, The Netherlands
| | - Bram L T Ramaekers
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre, P. Debyelaan 25, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
- Maastricht University, Care and Public Health Research Institute (CAPHRI), Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands
| | - Rogier Butter
- Department of Pathology, Cancer Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Joanne M Mankor
- Department of Pulmonary Medicine, Erasmus Medical Centre, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Marthe S Paats
- Department of Pulmonary Medicine, Erasmus Medical Centre, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Joachim G J V Aerts
- Department of Pulmonary Medicine, Erasmus Medical Centre, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Zakile A Mfumbilwa
- Department of Epidemiology and Data Science, Amsterdam University Medical Center-Location VUmc, Van der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands
| | - Paul Roepman
- Hartwig Medical Foundation, Science Park 408, 1098 XH, Amsterdam, The Netherlands
| | - Veerle M H Coupé
- Department of Epidemiology and Data Science, Amsterdam University Medical Center-Location VUmc, Van der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands
| | - Carin A Uyl-de Groot
- Erasmus School of Health Policy and Management/Institute for Medical Technology Assessment, Erasmus University Rotterdam, Burgemeester Oudlaan 50, 3062 PA, Rotterdam, The Netherlands
| | - Wim H van Harten
- Department of Psychosocial Research and Epidemiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
- Department of Health Technology and Services Research, University of Twente, Hallenweg 5, 7522 NH, Enschede, The Netherlands
| | - Manuela A Joore
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre, P. Debyelaan 25, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
- Maastricht University, Care and Public Health Research Institute (CAPHRI), Universiteitssingel 40, 6229 ER, Maastricht, The Netherlands.
| |
Collapse
|
28
|
The storm of NGS in NSCLC diagnostic-therapeutic pathway: How to sun the real clinical practice. Crit Rev Oncol Hematol 2021; 169:103561. [PMID: 34856311 DOI: 10.1016/j.critrevonc.2021.103561] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/18/2022] Open
Abstract
The increasing number of approved drugs along with next generation sequencing (NGS) technologies look out as potential revolution of biomolecular characterization of non-small-cell lung cancer (NSCLC). Nevertheless, several aspects impact on success rate of NGS in clinical practice: a multidisciplinary approach and thorough knowledge of strengths and limits of each technologic diagnostic tool are required. Crucial preliminary step is the selection of the best available sample before testing, aware of clinical condition and setting of disease. Genomic data should be than integrated in the clinical context and matched with available therapeutic options; Molecular Tumor Boards (MTB) are worldwide emerging interdisciplinary groups implemented to transfer the impact of precision medicine in clinical practice. In order to guarantee equity in treatment, these considerations should find their application widely and rapidly. Aim of this review is offering an overview of emerging biomarkers, relative upcoming targeted drugs, and new diagnostic chances with an authors' perspective about a real-life diagnostic-therapeutic algorithm useful for daily clinical practice.
Collapse
|
29
|
Devarakonda S, Li Y, Martins Rodrigues F, Sankararaman S, Kadara H, Goparaju C, Lanc I, Pepin K, Waqar SN, Morgensztern D, Ward J, Masood A, Fulton R, Fulton L, Gillette MA, Satpathy S, Carr SA, Wistuba I, Pass H, Wilson RK, Ding L, Govindan R. Genomic Profiling of Lung Adenocarcinoma in Never-Smokers. J Clin Oncol 2021; 39:3747-3758. [PMID: 34591593 PMCID: PMC8601276 DOI: 10.1200/jco.21.01691] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/20/2021] [Accepted: 08/24/2021] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Approximately 10%-40% of patients with lung cancer report no history of tobacco smoking (never-smokers). We analyzed whole-exome and RNA-sequencing data of 160 tumor and normal lung adenocarcinoma (LUAD) samples from never-smokers to identify clinically actionable alterations and gain insight into the environmental and hereditary risk factors for LUAD among never-smokers. METHODS We performed whole-exome and RNA-sequencing of 88 and 69 never-smoker LUADs. We analyzed these data in conjunction with data from 76 never-smoker and 299 smoker LUAD samples sequenced by The Cancer Genome Atlas and Clinical Proteomic Tumor Analysis Consortium. RESULTS We observed a high prevalence of clinically actionable driver alterations in never-smoker LUADs compared with smoker LUADs (78%-92% v 49.5%; P < .0001). Although a subset of never-smoker samples demonstrated germline alterations in DNA repair genes, the frequency of samples showing germline variants in cancer predisposing genes was comparable between smokers and never-smokers (6.4% v 6.9%; P = .82). A subset of never-smoker samples (5.9%) showed mutation signatures that were suggestive of passive exposure to cigarette smoke. Finally, analysis of RNA-sequencing data showed distinct immune transcriptional subtypes of never-smoker LUADs that varied in their expression of clinically relevant immune checkpoint molecules and immune cell composition. CONCLUSION In this comprehensive genomic and transcriptome analysis of never-smoker LUADs, we observed a potential role for germline variants in DNA repair genes and passive exposure to cigarette smoke in the pathogenesis of a subset of never-smoker LUADs. Our findings also show that clinically actionable driver alterations are highly prevalent in never-smoker LUADs, highlighting the need for obtaining biopsies with adequate cellularity for clinical genomic testing in these patients.
Collapse
Affiliation(s)
- Siddhartha Devarakonda
- Division of Oncology, Washington University School of Medicine, St Louis, MO
- Siteman Cancer Center, St Louis, MO
| | - Yize Li
- Division of Oncology, Washington University School of Medicine, St Louis, MO
- McDonnell Genome Institute, St Louis, MO
- Division of Biological and Biomedical Sciences, Washington University in St Louis, St Louis, MO
| | - Fernanda Martins Rodrigues
- Division of Oncology, Washington University School of Medicine, St Louis, MO
- McDonnell Genome Institute, St Louis, MO
- Division of Biological and Biomedical Sciences, Washington University in St Louis, St Louis, MO
| | | | | | | | | | - Kymberlie Pepin
- Division of Oncology, Washington University School of Medicine, St Louis, MO
| | - Saiama N. Waqar
- Division of Oncology, Washington University School of Medicine, St Louis, MO
- Siteman Cancer Center, St Louis, MO
| | - Daniel Morgensztern
- Division of Oncology, Washington University School of Medicine, St Louis, MO
- Siteman Cancer Center, St Louis, MO
| | - Jeffrey Ward
- Division of Oncology, Washington University School of Medicine, St Louis, MO
- Siteman Cancer Center, St Louis, MO
| | | | | | | | - Michael A. Gillette
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Shankha Satpathy
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | - Steven A. Carr
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
| | | | - Harvey Pass
- New York University Langone Medical Center, New York, NY
| | | | - Li Ding
- Division of Oncology, Washington University School of Medicine, St Louis, MO
- Siteman Cancer Center, St Louis, MO
- McDonnell Genome Institute, St Louis, MO
- Department of Genetics, Washington University in St Louis, St Louis, MO
- Department of Medicine, Washington University in St Louis, St Louis, MO
| | - Ramaswamy Govindan
- Division of Oncology, Washington University School of Medicine, St Louis, MO
- Siteman Cancer Center, St Louis, MO
| |
Collapse
|
30
|
Sheinson DM, Wong WB, Flores C, Ogale S, Gross CP. Association Between Medicare's National Coverage Determination and Utilization of Next-Generation Sequencing. JCO Oncol Pract 2021; 17:e1774-e1784. [PMID: 34043456 PMCID: PMC8600504 DOI: 10.1200/op.20.01023] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/18/2021] [Accepted: 04/26/2021] [Indexed: 12/30/2022] Open
Abstract
PURPOSE In 2018, Medicare issued a national coverage determination (NCD) providing reimbursement for next-generation sequencing (NGS) tests for beneficiaries with advanced or metastatic cancer and no previous NGS testing. We examined the association between NCD implementation and NGS utilization trends in Medicare beneficiaries versus commercially insured patients. METHODS This was a retrospective study of patients with advanced non-small-cell lung cancer (aNSCLC), metastatic colorectal cancer (mCRC), metastatic breast cancer (mBC), or advanced melanoma with a de novo or recurrent advanced diagnosis from January 1, 2011, through December 30, 2019, using a nationwide US electronic health record-derived deidentified database. Patients were classified by insurance and by advanced diagnosis date. NGS testing was assessed by receipt of first NGS test result ≤ 60 days of advanced diagnosis. Interrupted time series analysis assessed NGS utilization pre- and post-NCD effective date by insurance type. RESULTS The utilization and repeat NGS testing analysis included 70,290 and 4,295 patients, respectively. Use of NGS rose from < 1% in 2011 to > 45% in Q4 2019 in aNSCLC while remaining < 20% in mBC and advanced melanoma. Among patients with aNSCLC, mCRC, or mBC, NGS testing increased post-NCD versus pre-NCD (P < .05). There was no significant difference in trends pre- and post-NCD between Medicare beneficiaries and commercially insured patients in any tumor. Repeat NGS testing was similar before the NCD (Medicare v commercial: 24.8% v 28.5%). Post-NCD, fewer Medicare beneficiaries had repeat NGS testing (27.7% v 36.0%; P < .01). CONCLUSION Trends in NGS utilization significantly changed post-NCD, although the magnitude of change was not significantly different by insurance type, indicating private insurers may also be incorporating NCD guidance. Implementation of the NCD may have limited use of repeat NGS testing in Medicare beneficiaries.
Collapse
Affiliation(s)
| | | | | | | | - Cary P. Gross
- Cancer Outcomes Public Policy and Effectiveness Research (COPPER) Center, Yale Cancer Center, New Haven, CT
| |
Collapse
|
31
|
An analysis of research biopsy core variability from over 5000 prospectively collected core samples. NPJ Precis Oncol 2021; 5:94. [PMID: 34707215 PMCID: PMC8551285 DOI: 10.1038/s41698-021-00234-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/22/2021] [Indexed: 12/28/2022] Open
Abstract
Factors correlated with biopsy tissue adequacy and the prevalence of within-biopsy variability were evaluated. Totally, 1149 research biopsies were performed on 686 patients from which 5090 cores were assessed. Biopsy cores were reviewed for malignant percentage (estimated percentage of cells in the core that were malignant) and malignant area (estimated area occupied by malignant cells). Linear mixed models and generalized linear mixed models were used for the analysis. A total of 641 (55.8%) biopsies contained a core with <10% malignant percentage (inadequate core). The chance of an inadequate core was not influenced by core order, though the malignant area decreased with each consecutive core (p < 0.001). Younger age, bone biopsy location, appendiceal tumor pathology, and responding/stable disease prior to biopsy increased the odds of a biopsy containing zero adequate cores. Within-biopsy variability in core adequacy is prevalent and suggests the need for histological tumor quality assessment of each core in order to optimize translational analyses.
Collapse
|
32
|
Niogret J, Dalens L, Truntzer C, Chevrier S, Favier L, Lagrange A, Coudert B, Fraisse C, Foucher P, Zouak A, Westeel V, Goussot V, Dérangère V, Albuisson J, Arnould L, Boidot R, Kaderbhai CG, Ghiringhelli F. Does large NGS panel analysed using exome tumour sequencing improve the management of advanced non-small-cell lung cancers? Lung Cancer 2021; 161:98-107. [PMID: 34560426 DOI: 10.1016/j.lungcan.2021.08.013] [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: 10/08/2020] [Revised: 03/02/2021] [Accepted: 08/24/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Non-small-cell lung cancer (NSCLC) is one of the most common and deadly cancers. Several molecular drivers of oncogene addiction are now known to be strong predictive biomarkers for target therapies. Advances in large Next Generation Sequencing (LNGS) have improved the ability to detect potentially targetable mutations. However, the integration of LNGS into clinical management in an individualized manner remains challenging. METHODS In this single-center observational study we included all patients with advanced NSCLC who underwent LNGS. Somatic and germline exome analysis was performed with a restriction on 323 cancer related genes. Variants were classified and Molecular Tumour Board (MTB) made therapeutic propositions. RESULTS We performed LNGS analysis in 281 patients with advanced NSCLC between March 2015 and January 2018. Technical failure occurred in only 3% of cases. Three hundred and fifty-six targetable mutations were detected. At least one targetable mutation was found in 209 patients. For all these patients, the MTB was able to recommend treatment with a targeted agent based on the evaluation of the tumour's genetic profile and treatment history. Twenty-nine patients (13.9%) were subsequently treated with an MTB-recommended targeted therapy. We did not observe any improvement in terms of clinical benefit for these patients. CONCLUSIONS In this case series, we show that including LNGS into routine clinical management was feasible but does not appear to provide clinical benefit in the management of patients with advanced NSCLC.
Collapse
Affiliation(s)
- Julie Niogret
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; University of Burgundy-Franche Comté, Maison de l'université Esplanade Erasme, 21000 Dijon, France
| | - Lorraine Dalens
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; University of Burgundy-Franche Comté, Maison de l'université Esplanade Erasme, 21000 Dijon, France
| | - Caroline Truntzer
- Platform of Transfert in Biological Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; Genomic and Immunotherapy Medical Institute, Dijon University Hospital, 14 rue Paul Gaffarel 21000 Dijon, France
| | - Sandy Chevrier
- Platform of Transfert in Biological Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Laure Favier
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Aurélie Lagrange
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Bruno Coudert
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Cléa Fraisse
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Pascal Foucher
- Department of Thoracic Oncology, Dijon University Hospital, 14 rue Paul Gaffarel, 21000 Dijon, France
| | - Ayoub Zouak
- Department of Thoracic Oncology, Dijon University Hospital, 14 rue Paul Gaffarel, 21000 Dijon, France
| | - Virginie Westeel
- Department of Pneumology, Besançon University Hospital, 3 Boulevard Alexandre Fleming, 25000 Besançon, France
| | - Vincent Goussot
- Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Valentin Dérangère
- University of Burgundy-Franche Comté, Maison de l'université Esplanade Erasme, 21000 Dijon, France; Platform of Transfert in Biological Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; UMR INSERM 1231, 7 Boulevard Jeanne d'Arc, 21000 Dijon, France
| | - Juliette Albuisson
- Platform of Transfert in Biological Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; Genomic and Immunotherapy Medical Institute, Dijon University Hospital, 14 rue Paul Gaffarel 21000 Dijon, France; Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Laurent Arnould
- Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - Romain Boidot
- University of Burgundy-Franche Comté, Maison de l'université Esplanade Erasme, 21000 Dijon, France; Platform of Transfert in Biological Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; Genomic and Immunotherapy Medical Institute, Dijon University Hospital, 14 rue Paul Gaffarel 21000 Dijon, France; Department of Tumour Biology and Pathology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; UMR INSERM 1231, 7 Boulevard Jeanne d'Arc, 21000 Dijon, France
| | - Courèche-Guillaume Kaderbhai
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France
| | - François Ghiringhelli
- Department of Medical Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; University of Burgundy-Franche Comté, Maison de l'université Esplanade Erasme, 21000 Dijon, France; Platform of Transfert in Biological Oncology, Georges François Leclerc Cancer Center - UNICANCER, 1 rue du Professeur Marion, Dijon 21000, France; Genomic and Immunotherapy Medical Institute, Dijon University Hospital, 14 rue Paul Gaffarel 21000 Dijon, France; UMR INSERM 1231, 7 Boulevard Jeanne d'Arc, 21000 Dijon, France.
| |
Collapse
|
33
|
Next Generation Sequencing Technology in Lung Cancer Diagnosis. BIOLOGY 2021; 10:biology10090864. [PMID: 34571741 PMCID: PMC8467994 DOI: 10.3390/biology10090864] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 12/29/2022]
Abstract
Simple Summary Lung cancer is still one of the most commonly diagnosed and deadliest cancers in the world. Its diagnosis at an early stage is highly necessary and will improve the standard of care of this disease. The aim of this article is to review the importance and applications of next generation sequencing in lung cancer diagnosis. As observed in many studies, next generation sequencing has been proven as a very helpful tool in the early detection of different types of cancers, including lung cancer, and has been used in the clinic, mainly due to its many advantages, such as low cost, speed, efficacy, low quantity usage of biological samples, and diversity. Abstract Lung cancer is still one of the most commonly diagnosed cancers, and one of the deadliest. The high death rate is mainly due to the late stage of diagnosis and low response rate to therapy. Previous and ongoing research studies have tried to discover new reliable and useful cbiomarkers for the diagnosis and prognosis of lung cancer. Next generation sequencing has become an essential tool in cancer diagnosis, prognosis, and evaluation of the treatment response. This article aims to review the leading research and clinical applications in lung cancer diagnosis using next generation sequencing. In this scope, we identified the most relevant articles that present the successful use of next generation sequencing in identifying biomarkers for early diagnosis correlated to lung cancer diagnosis and treatment. This technique can be used to evaluate a high number of biomarkers in a short period of time and from small biological samples, which makes NGS the preferred technique to develop clinical tests for personalized medicine using liquid biopsy, the new trend in oncology.
Collapse
|
34
|
Kuijvenhoven JC, Kramer T, Korevaar DA, Ninaber MK, Trisolini R, Szlubowski A, Gnass M, von der Thüsen J, Cohen D, Bonta PI, Annema JT. Endobronchial ultrasound in diagnosing and staging of lung cancer by Acquire 22G TBNB versus regular 22G TBNA needles: study protocol of a randomised clinical trial. BMJ Open 2021; 11:e051820. [PMID: 34475187 PMCID: PMC8413963 DOI: 10.1136/bmjopen-2021-051820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Accurate diagnosis and staging of lung cancer is crucial because it directs treatment and prognosis. Endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) and endoscopic ultrasound with bronchoscope fine-needle aspiration (EUS-B-FNA) are important in this process by sampling hilar/mediastinal lymph nodes and centrally located lung tumours. With the upcoming of immunotherapy and targeted therapies, assessment of programmed death ligand 1 (PD-L1) expression and molecular profiling has become important but is often impossible in cytological samples obtained through standard 22G TBNA needles. Recently, a three-pronged cutting edge 22G needle was developed that allows for transbronchial needle biopsy (TBNB). Our objective is to determine if EBUS/EUS-B-guided nodal/lung tumour sampling with Acquire 22G TBNB needles results in an improved suitability rate for the assessment of PD-L1 expression in comparison to standard 22G TBNA needles in patients with a final diagnosis of lung cancer. METHODS AND ANALYSIS This is an investigator-initiated, parallel group randomised clinical trial. Patients are recruited at respiratory medicine outpatient clinics of participating university and general hospitals in the Netherlands, Poland and Italy. In total 158 adult patients with (suspected) lung cancer are included if they have an indication for mediastinal/hilar lymph node or lung tumour sampling by EBUS-TBNA and/or EUS-B-FNA based on current clinical guidelines. Web-based randomisation between the two needles will be performed. Samples obtained from mediastinal/hilar lymph nodes and/or primary tumour will be processed for cytology smears and cell block analysis and reviewed by blinded reference pathologists. An intention-to-treat analysis will be applied. Patients with missing data will be excluded from analysis for that specific variable but included in the analysis of other variables. This study is financially supported by Boston Scientific. ETHICS AND DISSEMINATION The study was approved by the local Ethics Committee (Medisch Ethische Toetsingscommissie Amsterdam Medical Center (AMC)). Dissemination will involve publication in a peer-reviewed biomedical journal. TRIAL REGISTRATION NUMBER NL7701; Pre-results.
Collapse
Affiliation(s)
- Jolanda C Kuijvenhoven
- Department of Respiratory Medicine, Amsterdam University Medical Centres, Amsterdam, The Netherlands
- Department of Respiratory Medicine, Medical Centre Leeuwarden, Leeuwarden, The Netherlands
| | - Tess Kramer
- Department of Respiratory Medicine, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Daniël A Korevaar
- Department of Respiratory Medicine, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Maarten K Ninaber
- Department of Respiratory Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Rocco Trisolini
- Department of Respiratory Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Artur Szlubowski
- Department of Respiratory Medicine, Pulmonary Hospital, Zakopane, Poland
| | - Maciej Gnass
- Department of Respiratory Medicine, Pulmonary Hospital, Zakopane, Poland
| | | | - Danielle Cohen
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter I Bonta
- Department of Respiratory Medicine, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| | - Jouke T Annema
- Department of Respiratory Medicine, Amsterdam University Medical Centres, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Devarakonda S, Pellini B, Verghese L, Park H, Morgensztern D, Govindan R, Suresh R, Oppelt P, Baggstrom MQ, Wu N, Waqar SN. A phase II study of everolimus in patients with advanced solid malignancies with TSC1, TSC2, NF1, NF2 or STK11 mutations. J Thorac Dis 2021; 13:4054-4062. [PMID: 34422335 PMCID: PMC8339787 DOI: 10.21037/jtd-21-195] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/28/2021] [Indexed: 11/06/2022]
Abstract
Background Activation of the mTOR pathway has been implicated in the development of several malignancies and alterations in TSC1, TSC2, STK11 and NF1, can lead to the dysregulation of this pathway. Furthermore, mutations in TSC1 and NF2 are known to confer sensitivity to everolimus-an mTOR inhibitor. Based on these data, a single-arm, open label, single-institution phase II basket study was designed to assess the activity of everolimus in patients with solid malignancies whose tumors harbored mutations in TSC1, TSC2, NF1, NF2, or STK11. Methods A total of 12 patients with histologically confirmed diagnosis of advanced solid tumors (metastatic, recurrent, or unresectable) with mutations in TSC1, TSC2, NF1, NF2 or STK11 genes, who had failed at least one line of standard of care systemic therapy, were enrolled to this open label, single-arm study. Presence of mutations in TSC1, TSC2, NF1, NF2 or STK11 genes was assessed using targeted-next generation sequencing (NGS). All eligible patients were treated with everolimus at an initial dose of 10 mg orally once daily in cycles of 28 days. The primary endpoint of this study was overall response rate (ORR). Results Of 12 patients enrolled, 8 were evaluable for response at the end of 2 cycles. One complete response (CR) was observed (12.5%) and one patient (12.5%) had stable disease (SD), while six (75%) patients showed disease progression. Everolimus was overall well tolerated with anemia, decreased neutrophil and lymphocyte counts, peripheral edema and hyperglycemia representing the most common adverse events. One patient discontinued treatment due to a treatment related grade 4 pericardial effusion. Both patients with CR or SD had a diagnosis of lung adenocarcinoma with NF1 or STK11 mutations, respectively. Conclusions Although this study failed to meet its prespecified ORR threshold for success of 30% or higher, exploratory analyses suggest potential activity for everolimus in a subset of patients with lung adenocarcinomas with STK11 or NF1 mutations. Further studies are necessary to systematically explore the clinical activity of everolimus, potentially as a combination therapy, in these patients.
Collapse
Affiliation(s)
- Siddhartha Devarakonda
- Division of Medical Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Bruna Pellini
- Department of Thoracic Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Luke Verghese
- Division of Medical Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Haeseong Park
- Division of Medical Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel Morgensztern
- Division of Medical Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ramaswamy Govindan
- Division of Medical Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rama Suresh
- Division of Medical Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Peter Oppelt
- Division of Medical Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Maria Q Baggstrom
- Division of Medical Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ningying Wu
- Biostatistics Shared Resource, Public Health Science Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Saiama N Waqar
- Division of Medical Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
36
|
Limaye S. Synchronized Tissue Acquisition Techniques for Novel Biomarker Discovery: Are You Ready to Waltz? JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2021; 4:168-169. [PMID: 35663103 PMCID: PMC9138434 DOI: 10.36401/jipo-21-x3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 05/01/2023]
Affiliation(s)
- Sewanti Limaye
- Department of Medical Oncology, Kokilaben Dhirubhai Ambani Hospital, Mumbai, India
| |
Collapse
|
37
|
Hur JY, Lee KY. Characteristics and Clinical Application of Extracellular Vesicle-Derived DNA. Cancers (Basel) 2021; 13:3827. [PMID: 34359729 PMCID: PMC8345206 DOI: 10.3390/cancers13153827] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/25/2021] [Accepted: 07/27/2021] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) carry RNA, proteins, lipids, and diverse biomolecules for intercellular communication. Recent studies have reported that EVs contain double-stranded DNA (dsDNA) and oncogenic mutant DNA. The advantage of EV-derived DNA (EV DNA) over cell-free DNA (cfDNA) is the stability achieved through the encapsulation in the lipid bilayer of EVs, which protects EV DNA from degradation by external factors. The existence of DNA and its stability make EVs a useful source of biomarkers. However, fundamental research on EV DNA remains limited, and many aspects of EV DNA are poorly understood. This review examines the known characteristics of EV DNA, biogenesis of DNA-containing EVs, methylation, and next-generation sequencing (NGS) analysis using EV DNA for biomarker detection. On the basis of this knowledge, this review explores how EV DNA can be incorporated into diagnosis and prognosis in clinical settings, as well as gene transfer of EV DNA and its therapeutic potential.
Collapse
Affiliation(s)
- Jae Young Hur
- Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Seoul 05030, Korea;
- Department of Pathology, Konkuk University Medical Center, Seoul 05030, Korea
| | - Kye Young Lee
- Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Seoul 05030, Korea;
- Department of Pulmonary Medicine, Konkuk University School of Medicine, Seoul 05030, Korea
| |
Collapse
|
38
|
Qvick A, Stenmark B, Carlsson J, Isaksson J, Karlsson C, Helenius G. Liquid biopsy as an option for predictive testing and prognosis in patients with lung cancer. Mol Med 2021; 27:68. [PMID: 34217228 PMCID: PMC8254966 DOI: 10.1186/s10020-021-00331-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The aim of this study was to investigate the clinical value of liquid biopsy as a primary source for variant analysis in lung cancer. In addition, we sought to characterize liquid biopsy variants and to correlate mutational load to clinical data. METHODS Circulating cell-free DNA was extracted from plasma from patients with lung cancer (n = 60) and controls with benign lung disease (n = 16). Variant analysis was performed using the AVENIO ctDNA Surveillance kit and the results were correlated to clinical and variant analysis data from tumor tissue or cytology retrieved from clinical routine diagnostics. RESULTS There were significantly more variants detected in lung cancer cases compared to controls (p = 0.011), but no difference between the histological subgroups of lung cancer was found (p = 0.465). Furthermore, significantly more variants were detected in patients with stage IIIb-IV disease compared to patients with stage I-IIIa (median 7 vs 4, p = 0.017). Plasma cfDNA mutational load was significantly associated with overall survival (p = 0.010). The association persisted when adjusted for stage and ECOG performance status (HR: 3.64, 95% CI 1.37-9.67, p = 0.009). Agreement between tumor and plasma samples significantly differed with stage; patients with stage IIIb-IV disease showed agreement in 88.2% of the cases with clinically relevant variants, compared to zero cases in stage I-IIIa (p = 0.004). Furthermore, one variant in EGFR, two in KRAS, and one in BRAF were detected in plasma but not in tumor samples. CONCLUSION This study concludes that in the vast majority of advanced NSCLC patients a reliable variant analysis can be performed using liquid biopsy from plasma. Furthermore, we found that the number of variants in plasma is associated with prognosis, possibly indicating a strategy for closer follow up on this crucial patient group.
Collapse
Affiliation(s)
- Alvida Qvick
- Dept. of Laboratory Medicine, Örebro University Hospital, Södra Grev. Roseng., 701 85 Örebro, Sweden
- Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Bianca Stenmark
- Dept. of Laboratory Medicine, Örebro University Hospital, Södra Grev. Roseng., 701 85 Örebro, Sweden
- Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Jessica Carlsson
- Dept. of Urology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Johan Isaksson
- Dept. of Respiratory Medicine, Gävle Hospital, Gävle, Sweden
- Centre for Research and Development Region Gävleborg/Uppsala University, Gävle, Sweden
- Dept. of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | | - Gisela Helenius
- Dept. of Laboratory Medicine, Örebro University Hospital, Södra Grev. Roseng., 701 85 Örebro, Sweden
- Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
39
|
Moore A, Bar Y, Maurice-Dror C, Finkel I, Goldvaser H, Dudnik E, Goldstein DA, Gordon N, Billan S, Gutfeld O, Wolf I, Popovtzer A. Next-generation sequencing in thyroid cancers: do targetable alterations lead to a therapeutic advantage?: A multicenter experience. Medicine (Baltimore) 2021; 100:e26388. [PMID: 34160418 PMCID: PMC8238320 DOI: 10.1097/md.0000000000026388] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 04/29/2021] [Accepted: 05/25/2021] [Indexed: 01/04/2023] Open
Abstract
ABSTRACT Radioiodine-refractory thyroid cancers (IRTCs) are uncommon and have a poor prognosis. Treatment options for radioiodine-refractory and anaplastic tumors (ATCs) are limited. Although the genomic landscape of thyroid cancer has been studied, there is little evidence on whether next-generation sequencing (NGS) findings translate to tumor control.We analyzed all patients with IRTC and ATC who underwent commercially available NGS in 3 cancer centers.Twenty-two patients were identified, 16 patients with IRTCs and 6 patients with ATCs. Eighteen (82%) had targetable findings in NGS, nine patients were treated accordingly. Median progression-free survival for targeted treatment was 50 months [95% confidence interval (CI95%) 9.8-66.6] and2 months (CI95% 0.2-16.5) for IRTC and ATC, respectively. Of 4 patients who achieved durable responses of 7 to 50 months, 2 are ongoing. The estimated median OS of IRTC receiving targeted treatment was not reached (CI95% 89.7-111.4 months) and was 77.8 months (CI95% 52.5-114.6) for patients treated conventionally (P = .3).NGS may detect clinically significant genetic alterations and benefit patients with advanced thyroid cancers.
Collapse
Affiliation(s)
- Assaf Moore
- Institute of Oncology, Davidoff Center, Rabin Medical Center – Beilinson Hospital, Petach Tikva
- Sackler Faculty of Medicine, Tel Aviv University
| | - Yael Bar
- Sackler Faculty of Medicine, Tel Aviv University
- Oncology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv
| | - Corinne Maurice-Dror
- Institute of Oncology, Rambam Health Care Campus
- Ruth & Bruce Rappaport, Faculty of Medicine, Technion Israel Institute of Technology, Haifa
| | - Inbar Finkel
- Institute of Oncology, Davidoff Center, Rabin Medical Center – Beilinson Hospital, Petach Tikva
| | - Hadar Goldvaser
- Oncology Institute, Shaare Zedek Medical Center
- The Hebrew University Faculty of Medicine, Jerusalem, Israel
| | - Elizabeth Dudnik
- Institute of Oncology, Davidoff Center, Rabin Medical Center – Beilinson Hospital, Petach Tikva
- Sackler Faculty of Medicine, Tel Aviv University
| | - Daniel A. Goldstein
- Institute of Oncology, Davidoff Center, Rabin Medical Center – Beilinson Hospital, Petach Tikva
- Sackler Faculty of Medicine, Tel Aviv University
| | - Noa Gordon
- Institute of Oncology, Davidoff Center, Rabin Medical Center – Beilinson Hospital, Petach Tikva
| | - Salem Billan
- Institute of Oncology, Rambam Health Care Campus
- Ruth & Bruce Rappaport, Faculty of Medicine, Technion Israel Institute of Technology, Haifa
| | - Orit Gutfeld
- Sackler Faculty of Medicine, Tel Aviv University
- Oncology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv
| | - Ido Wolf
- Sackler Faculty of Medicine, Tel Aviv University
- Oncology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv
| | - Aron Popovtzer
- Institute of Oncology, Davidoff Center, Rabin Medical Center – Beilinson Hospital, Petach Tikva
- Sackler Faculty of Medicine, Tel Aviv University
| |
Collapse
|
40
|
Fukui T, Sakai K, Sasaki J, Kakegawa MI, Igawa S, Mitsufuji H, Takeda M, Takahama T, Nakagawa K, Nishio K, Naoki K. Implementation of clinical sequencing for molecular profiling in patients with advanced cancer. Cancer Biomark 2021; 31:119-126. [PMID: 33896820 DOI: 10.3233/cbm-200781] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND The advancement of cancer genomics has allowed for multiplex gene assays using next-generation sequencing (NGS) to be practically implemented, however, a clinical practice system remains to be established. OBJECTIVE We evaluated the feasibility of clinical sequencing using NGS-based multiplex gene assays between cooperating medical institutions in patients with advanced cancers. METHODS In this observational study, DNA and RNA samples prepared from existing tumor tissues were subjected to comprehensive genomic profiling using targeted sequencing. RESULTS From January 2017 to March 2019, 36 samples from 33 patients were assessed. Of all patients, 27 (82%) had lung cancer, with the median age of 50 years (range 38-83). Multiplex gene panel tests were successfully carried out on 35/36 (97%) samples. Potentially actionable gene alterations were identified in 10/30 (33%) samples (3 HER2, 2 KRAS, 2 ALK, 1 PIK3CA, 1 RET, and 1 CDKN2A). In the 6 samples examined for resistant mechanisms, ALK I1171N mutation and MET copy number gain were detected in 2 patients with ALK rearrangement-positive lung cancer. CONCLUSIONS Clinical sequencing using NGS-based multiplex gene assays between collaborating domestic medical institutions was feasible, with a success rate of > 97%. Overall, clinical sequencing benefits therapeutic decision-making in patients with advanced cancer.
Collapse
Affiliation(s)
- Tomoya Fukui
- Department of Respiratory Medicine, Kitasato University School of Medicine, Kanagawa, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Jiichiro Sasaki
- Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, Kanagawa, Japan
| | | | - Satoshi Igawa
- Department of Respiratory Medicine, Kitasato University School of Medicine, Kanagawa, Japan
| | - Hisashi Mitsufuji
- Fundamental Nursing, Kitasato University School of Nursing, Kanagawa, Japan
| | - Masayuki Takeda
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Takayuki Takahama
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Katsuhiko Naoki
- Department of Respiratory Medicine, Kitasato University School of Medicine, Kanagawa, Japan
| |
Collapse
|
41
|
Rawal S, Patel M. Bio-Nanocarriers for Lung Cancer Management: Befriending the Barriers. NANO-MICRO LETTERS 2021; 13:142. [PMID: 34138386 PMCID: PMC8196938 DOI: 10.1007/s40820-021-00630-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/23/2021] [Indexed: 05/03/2023]
Abstract
Lung cancer is a complex thoracic malignancy developing consequential to aberrations in a myriad of molecular and biomolecular signaling pathways. It is one of the most lethal forms of cancers accounting to almost 1.8 million new annual incidences, bearing overall mortality to incidence ratio of 0.87. The dismal prognostic scenario at advanced stages of the disease and metastatic/resistant tumor cell populations stresses the requisite of advanced translational interdisciplinary interventions such as bionanotechnology. This review article deliberates insights and apprehensions on the recent prologue of nanobioengineering and bionanotechnology as an approach for the clinical management of lung cancer. The role of nanobioengineered (bio-nano) tools like bio-nanocarriers and nanobiodevices in secondary prophylaxis, diagnosis, therapeutics, and theranostics for lung cancer management has been discussed. Bioengineered, bioinspired, and biomimetic bio-nanotools of considerate translational value have been reviewed. Perspectives on existent oncostrategies, their critical comparison with bio-nanocarriers, and issues hampering their clinical bench side to bed transformation have also been summarized.
Collapse
Affiliation(s)
- Shruti Rawal
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad, Gujarat, 382 481, India
| | - Mayur Patel
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad, Gujarat, 382 481, India.
| |
Collapse
|
42
|
Choi YJ, Choi JY, Kim JW, Lim AR, Lee Y, Chang WJ, Lee S, Sung JS, Chung HJ, Lee JW, Kang EJ, Kim JS, Lim T, Kim HS, Kim YJ, Ahn MS, Kim YS, Park JH, Lim S, Cho SS, Cho JH, Shin SW, Park KH, Kim YH. Comparison of the Data of a Next-Generation Sequencing Panel from K-MASTER Project with that of Orthogonal Methods for Detecting Targetable Genetic Alterations. Cancer Res Treat 2021; 54:30-39. [PMID: 34015890 PMCID: PMC8756135 DOI: 10.4143/crt.2021.218] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/18/2021] [Indexed: 11/21/2022] Open
Abstract
Purpose K-MASTER project is a Korean national precision medicine platform that screened actionable mutations by analyzing next-generation sequencing (NGS) of solid tumor patients. We compared gene analyses between NGS panel from the K-MASTER project and orthogonal methods. Materials and Methods Colorectal, breast, non–small cell lung, and gastric cancer patients were included. We compared NGS results from K-MASTER projects with those of non-NGS orthogonal methods (KRAS, NRAS, and BRAF mutations in colorectal cancer [CRC]; epidermal growth factor receptor [EGFR], anaplastic lymphoma kinase [ALK] fusion, and reactive oxygen species 1 [ROS1] fusion in non–small cell lung cancer [NSCLC], and Erb-B2 receptor tyrosine kinase 2 (ERBB2) positivity in breast and gastric cancers). Results In the CRC cohort (n=225), the sensitivity and specificity of NGS were 87.4% and 79.3% (KRAS); 88.9% and 98.9% (NRAS); and 77.8% and 100.0% (BRAF), respectively. In the NSCLC cohort (n=109), the sensitivity and specificity of NGS for EGFR were 86.2% and 97.5%, respectively. The concordance rate for ALK fusion was 100%, but ROS1 fusion was positive in only one of three cases that were positive in orthogonal tests. In the breast cancer cohort (n=260), ERBB2 amplification was detected in 45 by NGS. Compared with orthogonal methods that integrated immunohistochemistry and in situ hybridization, sensitivity and specificity were 53.7% and 99.4%, respectively. In the gastric cancer cohort (n=64), ERBB2 amplification was detected in six by NGS. Compared with orthogonal methods, sensitivity and specificity were 62.5% and 98.2%, respectively. Conclusion The results of the K-MASTER NGS panel and orthogonal methods showed a different degree of agreement for each genetic alteration, but generally showed a high agreement rate.
Collapse
Affiliation(s)
- Yoon Ji Choi
- Division of Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea.,Cancer Precision Medicine Diagnosis and Treatment Enterprise, Korea University Anam Hospital, Seoul, Korea
| | - Jung Yoon Choi
- Division of Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea.,Cancer Precision Medicine Diagnosis and Treatment Enterprise, Korea University Anam Hospital, Seoul, Korea
| | - Ju Won Kim
- Division of Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea.,Cancer Precision Medicine Diagnosis and Treatment Enterprise, Korea University Anam Hospital, Seoul, Korea
| | - Ah Reum Lim
- Division of Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea.,Cancer Precision Medicine Diagnosis and Treatment Enterprise, Korea University Anam Hospital, Seoul, Korea
| | - Youngwoo Lee
- Division of Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Won Jin Chang
- Division of Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea.,Cancer Precision Medicine Diagnosis and Treatment Enterprise, Korea University Anam Hospital, Seoul, Korea
| | - Soohyeon Lee
- Division of Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea.,Cancer Precision Medicine Diagnosis and Treatment Enterprise, Korea University Anam Hospital, Seoul, Korea
| | - Jae Sook Sung
- Cancer Precision Medicine Diagnosis and Treatment Enterprise, Korea University Anam Hospital, Seoul, Korea
| | - Hee-Joon Chung
- Cancer Precision Medicine Diagnosis and Treatment Enterprise, Korea University Anam Hospital, Seoul, Korea
| | - Jong Won Lee
- Brain Korea 21 Plus Project for Biomedical Science, Korea University College of Medicine, Seoul, Korea
| | - Eun Joo Kang
- Division of Oncology, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea
| | - Jung Sun Kim
- Division of Oncology, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea
| | - Taekyu Lim
- Division of Hematology-Oncology, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, Korea
| | - Hye Sook Kim
- Department of Internal Medicine, Inje University Ilsan Hospital, Goyang, Korea
| | - Yu Jung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Mi Sun Ahn
- Ajou University Medical Center, Suwon, Korea
| | - Young Saing Kim
- Division of Medical Oncology, Department of Internal Medicine, Gil Medical Center, Gachon University College of Medicine, Incheon, Korea
| | - Ji Hyun Park
- Department of Hemato-Oncology, Konkuk Medical Center, University of Konkuk College of Medicine, Seoul, Korea
| | - Seungtaek Lim
- Department of Oncology, Wonju Severance Christianity Hospital, Wonju, Korea
| | - Sung Shim Cho
- Division of Medical Oncology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Jang Ho Cho
- Division of Oncology, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea
| | - Sang Won Shin
- Division of Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Kyong Hwa Park
- Division of Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea.,Cancer Precision Medicine Diagnosis and Treatment Enterprise, Korea University Anam Hospital, Seoul, Korea
| | - Yeul Hong Kim
- Division of Oncology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea.,Cancer Precision Medicine Diagnosis and Treatment Enterprise, Korea University Anam Hospital, Seoul, Korea
| |
Collapse
|
43
|
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: 1.3] [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
|
44
|
Zheng S, Wang X, Fu Y, Li B, Xu J, Wang H, Huang Z, Xu H, Qiu Y, Shi Y, Li K. Targeted next-generation sequencing for cancer-associated gene mutation and copy number detection in 206 patients with non-small-cell lung cancer. Bioengineered 2021; 12:791-802. [PMID: 33629637 PMCID: PMC8291840 DOI: 10.1080/21655979.2021.1890382] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The knowledge of genetic variation in Chinese patients with non–small-cell lung cancer (NSCLC) is still limited. We aimed to profile this genetic variation in 206 Chinese patients with NSCLC using next-generation sequencing. Tumor tissues or whole-blood samples were collected and subjected to whole-exome targeted next-generation sequencing, which included 565 tumor-associated genes, for somatic gene mutation screening and copy number variation (CNV) detection. Potential functions of most commonly mutated genes and genes with CNV were predicted by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Atotal of 18,749 mutations were identified using targeted next-generation sequencing, and 85.3% of them were missense mutations. Among the mutation, conversions between pyrimidine and purine were predominant, and C> T/G > A was the most common substitution type. High frequencies of mutations were noted in TP53 (47.6%), EGFR (41.7%), CREBBP (23.1%), KMT2C (16.9%), MUC2 (16.6%), DNMT3A (15.5%), LRP1B (15.5%), MUC4 (15.5%), CDC27 (15.2%), and KRAS (12.8%). EGFR and KRAS mutations were mutually exclusive. The tumor mutation load showed differences depending on gender and tumor type. CNV analysis showed that BCORL1 and ARAF have the highest copy number amplification, whereas KDM6A and RBM10 showed the highest copy number deletion. GO and KEGG analyses indicated that high-frequency mutations and CNV genes were concentrated in tumor-related PI3K-Akt, FoxO, and Ras signaling pathway. Cumulatively, we studied somatic gene mutations involved in NSCLC and predicted their clinical significance in Chinese population. These findings may provide clues for etiology and drug target of NSCLC.
Collapse
Affiliation(s)
- Songbai Zheng
- Translational Medicine Research Institute, Guangzhou Huayin Medical Laboratory Center Co., Ltd., Guangzhou, China
| | - Xiaodan Wang
- Translational Medicine Research Institute, Guangzhou Huayin Medical Laboratory Center Co., Ltd., Guangzhou, China
| | - Ying Fu
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,Research and Development Institute, Sinotech Genomics, Shanghai, China
| | - Beibei Li
- Laboratory Medicine Center, Shunde Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Laboratory Medicine Center, The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianhua Xu
- Laboratory Medicine Center, Shunde Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haifang Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhen Huang
- Translational Medicine Research Institute, Guangzhou Huayin Medical Laboratory Center Co., Ltd., Guangzhou, China
| | - Hui Xu
- Technical Service Department, Guangzhou Huayin Medical Research Institute Co., Ltd., Guangzhou, China
| | - Yurong Qiu
- Translational Medicine Research Institute, Guangzhou Huayin Medical Laboratory Center Co., Ltd., Guangzhou, China
| | - Yaozhou Shi
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.,CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Kui Li
- Translational Medicine Research Institute, Guangzhou Huayin Medical Laboratory Center Co., Ltd., Guangzhou, China.,Technical Service Department, Guangzhou Huayin Medical Research Institute Co., Ltd., Guangzhou, China
| |
Collapse
|
45
|
Siemanowski J, Heydt C, Merkelbach-Bruse S. Predictive molecular pathology of lung cancer in Germany with focus on gene fusion testing: Methods and quality assurance. Cancer Cytopathol 2021; 128:611-621. [PMID: 32885916 DOI: 10.1002/cncy.22293] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/29/2020] [Accepted: 05/04/2020] [Indexed: 12/18/2022]
Abstract
Predictive molecular testing has become an important part of the diagnosis of any patient with lung cancer. Using reliable methods to ensure timely and accurate results is inevitable for guiding treatment decisions. In the past few years, parallel sequencing has been established for mutation testing, and its use is currently broadened for the detection of other genetic alterations, such as gene fusion and copy number variations. In addition, conventional methods such as immunohistochemistry and in situ hybridization are still being used, either for formalin-fixed, paraffin-embedded tissue or for cytological specimens. For the development and broad implementation of such complex technologies, interdisciplinary and regional networks are needed. The Network Genomic Medicine (NGM) has served as a model of centralized testing and decentralized treatment of patients and incorporates all German comprehensive cancer centers. Internal quality control, laboratory accreditation, and participation in external quality assessment is mandatory for the delivery of reliable results. Here, we provide a summary of current technologies used to identify patients who have lung cancer with gene fusions, briefly describe the structures of NGM and the national NGM (nNGM), and provide recommendations for quality assurance.
Collapse
Affiliation(s)
- Janna Siemanowski
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Carina Heydt
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | | |
Collapse
|
46
|
Lee SE, Park HY, Hur JY, Kim HJ, Kim IA, Kim WS, Lee KY. Genomic profiling of extracellular vesicle-derived DNA from bronchoalveolar lavage fluid of patients with lung adenocarcinoma. Transl Lung Cancer Res 2021; 10:104-116. [PMID: 33569297 PMCID: PMC7867756 DOI: 10.21037/tlcr-20-888] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background Extracellular vesicles (EVs) are membrane-bound and nanometer-sized particles released from most types of cells, containing double-stranded DNA reflecting mutational status of the parental tumor cells. Furthermore, epidermal growth factor receptor (EGFR) genotyping using EV-derived DNA (EV DNA) in bronchoalveolar lavage fluid (BALF) showed almost 100% sensitivity in patients with advanced non-small cell lung cancer (NSCLC). Methods We assessed the technical performance of DNA derived from BALF-EV (BALF EV DNA) in targeted next-generation sequencing (NGS) for detection and quantification of mutations compared with the matching tissue DNA in 20 lung adenocarcinomas. Results DNA yields, tumor purity, and depth of coverage were higher using the tissue DNA than using the BALF EV DNA. However, estimated library size was not significantly different between the two samples, and BALF EV DNA yielded longer fragments than tissue DNA. Overall mutation concordance between the two samples were 56% for nonsynonymous somatic mutations and increased to 81% for clinically significant mutations. By-variant sensitivity for clinically significant somatic mutations increased from 62% to 83% in the NGS of BALF EV DNA. Allele frequencies of EGFR and TP53 were higher in tissue DNA (10–25%) than in BALF EV DNA (<5%). Tumor mutation burden of BALF EV DNA correlated with that of tissue DNA. Conclusions Our findings demonstrate, for the first time, that BALF EV DNA in patients with NSCLC can be a reliable DNA source for targeted NGS for the identification of actionable genetic alterations and that this approach has high clinical feasibility and utility.
Collapse
Affiliation(s)
- Seung Eun Lee
- Department of Pathology, Konkuk University School of Medicine, Seoul, Korea
| | - Ha Young Park
- Department of Pathology, Busan Paik Hospital, Inje University College of Medicine, Gimhae, Korea
| | - Jae Young Hur
- Department of Pathology, Konkuk University School of Medicine, Seoul, Korea.,Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Seoul, Korea
| | - Hee Joung Kim
- Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Seoul, Korea.,Department of Internal Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - In Ae Kim
- Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Seoul, Korea.,Department of Internal Medicine, Konkuk University School of Medicine, Seoul, Korea
| | - Wan Seop Kim
- Department of Pathology, Konkuk University School of Medicine, Seoul, Korea
| | - Kye Young Lee
- Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Seoul, Korea.,Department of Internal Medicine, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
47
|
Palmero R, Taus A, Viteri S, Majem M, Carcereny E, Garde-Noguera J, Felip E, Nadal E, Malfettone A, Sampayo M, Riva F, Nagy RJ, Lanman RB, Faull I, Dix D, Karachaliou N, Rosell R. Biomarker Discovery and Outcomes for Comprehensive Cell-Free Circulating Tumor DNA Versus Standard-of-Care Tissue Testing in Advanced Non–Small-Cell Lung Cancer. JCO Precis Oncol 2021; 5:93-102. [DOI: 10.1200/po.20.00241] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Treatment guidelines for advanced non–small-cell lung cancer (aNSCLC) recommend broad molecular profiling for targeted therapy selection. This study prospectively assessed comprehensive next-generation sequencing (NGS) of cell-free circulating tumor DNA (cfDNA) compared with standard-of-care (SOC) tissue-based testing to identify guideline-recommended alterations in aNSCLC. PATIENTS AND METHODS Patients with treatment-naïve aNSCLC were tested using a well-validated NGS cfDNA panel, and results were compared with SOC tissue testing. The primary objective was noninferiority of cfDNA vs. tissue analysis for the detection of two guideline-recommended biomarkers ( EGFR and ALK) and an additional six actionable biomarkers. Secondary analyses included tissue versus cfDNA biomarker discovery, overall response rate (ORR), progression-free survival (PFS) to targeted therapy, and positive predictive value (PPV) of cfDNA. RESULTS The primary objective was met with cfDNA identifying actionable mutations in 46 patients versus 48 by tissue ( P < .05). In total, 0/186 patients were genotyped for all eight biomarkers with tissue, compared with 90.8% using cfDNA. Targetable alterations or KRAS were identified in 80.7% when cfDNA was used first versus 57.1% when tissue was used first. PPV for cfDNA-detected EGFR was 100.0% (25/25). ORR and PFS in patients receiving targeted therapy based on tissue or cfDNA were similar to those previously reported. Conclusion This prospective study confirms a previous report that comprehensive cfDNA testing is noninferior to SOC tissue testing in detecting aNSCLC-recommended biomarkers. Furthermore, cfDNA-based first-line therapy produced outcomes similar to tissue-based testing, demonstrating the clinical utility of comprehensive cfDNA genotyping as the initial genotyping modality in patients with treatment-naïve aNSCLC when tissue is insufficient or when all actionable biomarkers cannot be rapidly assessed.
Collapse
Affiliation(s)
| | - Alvaro Taus
- Hospital del Mar, Barcelona, Spain
- Universidad Autónoma de Barcelona (UAB), Barcelona, Spain
| | - Santiago Viteri
- Quirón Salud-Dexeus University Institute, IOR, Medical Oncology Department, Barcelona, Spain
| | | | - Enric Carcereny
- Institut Català d'Oncologia Badalona-Hospital Germans Trias i Pujol B-ARGO, Badalona, Spain
| | | | | | | | - Andrea Malfettone
- Medica Scientia Innovation Research—MEDSIR, Barcelona, Spain and Ridgewood, NJ
| | - Miguel Sampayo
- Medica Scientia Innovation Research—MEDSIR, Barcelona, Spain and Ridgewood, NJ
| | - François Riva
- Medica Scientia Innovation Research—MEDSIR, Barcelona, Spain and Ridgewood, NJ
| | | | | | - Iris Faull
- Guardant Health, South San Francisco, CA
| | - Daniel Dix
- Guardant Health, South San Francisco, CA
| | - Niki Karachaliou
- Catalan Institute of Oncology, Hospital Germans Trias I Pujol, Badalona, Spain
| | - Rafael Rosell
- Catalan Institute of Oncology, Hospital Germans Trias I Pujol, Badalona, Spain
| |
Collapse
|
48
|
Dupain C, Masliah‐Planchon J, Gu C, Girard E, Gestraud P, Du Rusquec P, Borcoman E, Bello D, Ricci F, Hescot S, Sablin M, Tresca P, de Moura A, Loirat D, Frelaut M, Vincent‐Salomon A, Lecerf C, Callens C, Antonio S, Franck C, Mariani O, Bièche I, Kamal M, Le Tourneau C, Servois V. Fine-needle aspiration as an alternative to core needle biopsy for tumour molecular profiling in precision oncology: prospective comparative study of next-generation sequencing in cancer patients included in the SHIVA02 trial. Mol Oncol 2021; 15:104-115. [PMID: 32750212 PMCID: PMC7782085 DOI: 10.1002/1878-0261.12776] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/07/2020] [Accepted: 07/30/2020] [Indexed: 12/15/2022] Open
Abstract
High-throughput molecular profiling of solid tumours using core needle biopsies (CNB) allows the identification of actionable molecular alterations, with around 70% success rate. Although several studies have demonstrated the utility of small biopsy specimens for molecular testing, there remains debate as to the sensitivity of the less invasive fine-needle aspiration (FNA) compared to CNB to detect molecular alterations. We aimed to prospectively evaluate the potential of FNA to detect such alterations in various tumour types as compared to CNB in cancer patients included in the SHIVA02 trial. An in-house amplicon-based targeted sequencing panel (Illumina TSCA 99.3 kb panel covering 87 genes) was used to identify pathogenic variants and gene copy number variations (CNV) in concomitant CNB and FNA samples obtained from 61 patients enrolled in the SHIVA02 trial (NCT03084757). The main tumour types analysed were breast (38%), colon (15%), pancreas (11%), followed by cervix and stomach (7% each). We report 123 molecular alterations (85 variants, 23 amplifications and 15 homozygous deletions) among which 98 (80%) were concordant between CNB and FNA. The remaining discordances were mainly related to deletions status, yet undetected alterations were not exclusively specific to FNA. Comparative analysis of molecular alterations in CNB and FNA showed high concordance in terms of variants as well as CNVs identified. We conclude FNA could therefore be used in routine diagnostics workflow and clinical trials for tumour molecular profiling with the advantages of being minimally invasive and preserve tissue material needed for diagnostic, prognostic or theranostic purposes.
Collapse
Affiliation(s)
- Célia Dupain
- Department of Drug Development and Innovation (D3i)Institut CurieParis & Saint‐CloudFrance
| | | | - Céline Gu
- Department of PathologyInstitut CuriePSL Research UniversityParisFrance
| | - Elodie Girard
- INSERM U900 Research UnitInstitut CurieSaint‐CloudFrance
| | | | - Pauline Du Rusquec
- Department of Drug Development and Innovation (D3i)Institut CurieParis & Saint‐CloudFrance
| | - Edith Borcoman
- Department of Drug Development and Innovation (D3i)Institut CurieParis & Saint‐CloudFrance
| | - Diana Bello
- Department of Drug Development and Innovation (D3i)Institut CurieParis & Saint‐CloudFrance
| | - Francesco Ricci
- Department of Drug Development and Innovation (D3i)Institut CurieParis & Saint‐CloudFrance
| | - Ségolène Hescot
- Department of Drug Development and Innovation (D3i)Institut CurieParis & Saint‐CloudFrance
| | - Marie‐Paule Sablin
- Department of Drug Development and Innovation (D3i)Institut CurieParis & Saint‐CloudFrance
| | - Patricia Tresca
- Department of Drug Development and Innovation (D3i)Institut CurieParis & Saint‐CloudFrance
| | - Alexandre de Moura
- Department of Drug Development and Innovation (D3i)Institut CurieParis & Saint‐CloudFrance
| | - Delphine Loirat
- Department of Drug Development and Innovation (D3i)Institut CurieParis & Saint‐CloudFrance
| | - Maxime Frelaut
- Department of Drug Development and Innovation (D3i)Institut CurieParis & Saint‐CloudFrance
| | | | - Charlotte Lecerf
- Department of Drug Development and Innovation (D3i)Institut CurieParis & Saint‐CloudFrance
| | - Céline Callens
- Department of GeneticsInstitut CuriePSL Research UniversityParisFrance
| | - Samantha Antonio
- Department of GeneticsInstitut CuriePSL Research UniversityParisFrance
| | - Coralie Franck
- Department of GeneticsInstitut CuriePSL Research UniversityParisFrance
| | - Odette Mariani
- Department of PathologyInstitut CuriePSL Research UniversityParisFrance
| | - Ivan Bièche
- Department of GeneticsInstitut CuriePSL Research UniversityParisFrance
- INSERM U1016Faculty of Pharmaceutical and Biological SciencesParis Descartes UniversityParisFrance
| | - Maud Kamal
- Department of Drug Development and Innovation (D3i)Institut CurieParis & Saint‐CloudFrance
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i)Institut CurieParis & Saint‐CloudFrance
- INSERM U900 Research UnitInstitut CurieSaint‐CloudFrance
- Paris‐Saclay UniversityParisFrance
| | - Vincent Servois
- Department of RadiologyInstitut CuriePSL Research UniversityParis & Saint‐CloudFrance
| |
Collapse
|
49
|
Nemoto D, Yokose T, Katayama K, Murakami S, Kato T, Saito H, Suzuki M, Eriguchi D, Samejima J, Nagashima T, Ito H, Yamada K, Nakayama H, Masuda M. Tissue surface area and tumor cell count affect the success rate of the Oncomine Dx Target Test in the analysis of biopsy tissue samples. Thorac Cancer 2020; 12:194-200. [PMID: 33185331 PMCID: PMC7812068 DOI: 10.1111/1759-7714.13743] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/25/2022] Open
Abstract
Background The Oncomine Dx Target Test (ODxTT) is a next‐generation sequencing‐based companion diagnostic test which has been recently developed; however, its analysis success rate could be improved, especially for small samples. The aim of this study was to identify the pathological factors associated with biopsy specimens that affect the analysis success rate of ODxTT. Methods We retrospectively investigated 119 cases subjected to ODxTT at Kanagawa Cancer Center. Data pertaining to the results of BRAF V600E mutation analysis in ODxTT and pathological factors based on microscope slides were collected. Pathological factors including tissue surface area, tumor cell count, and tumor content rate were assessed. We constructed receiver operating characteristic curves and determined the optimal cutoff values of each pathological factor. Multivariate logistic analysis was used to identify significant factors. Results A total of 98 of 119 samples were successfully analyzed (75.6%). The tissue surface area and tumor cell count were significantly higher in the group associated with analysis success (P < 0.001 and P = 0.011, respectively), and their optimal cutoff values were 1.04 mm2 and 375 cells, respectively. A tissue surface area > 1.04 mm2 and tumor cell count >375 cells had a positive effect on the analysis success rate of ODxTT (odds ratio [OR] 0.10; 95% confidence interval [CI]: 0.03–0.35; P < 0.001 and OR 0.25; 95% CI: 0.07–0.90; P = 0.033, respectively). Conclusions Selecting samples with a tissue surface area > 1.04 mm2 and a tumor cell count >375 cells might improve the analysis success rate of ODxTT. Key points Significant findings of the study: We found that a tissue surface area > 1.04 mm2 and tumor cell count >375 cells had a positive effect on the analysis success rate of ODxTT in the analysis of biopsy tissue samples. What this study adds: It is sometimes necessary to assess genetic alterations with a small biopsy sample in daily practice. The criteria mentioned above will help to determine which tests should be performed, ODxTT or multiple single‐gene testing.
Collapse
Affiliation(s)
- Daiji Nemoto
- Department of Thoracic Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Tomoyuki Yokose
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Kayoko Katayama
- Unit of Cancer Survivorship and Education, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Shuji Murakami
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Terufumi Kato
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Haruhiro Saito
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Masaki Suzuki
- Department of Pathology, Kanagawa Cancer Center, Yokohama, Japan
| | - Daisuke Eriguchi
- Department of Thoracic Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Joji Samejima
- Department of Thoracic Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Takuya Nagashima
- Department of Thoracic Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Hiroyuki Ito
- Department of Thoracic Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Kouzo Yamada
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Haruhiko Nakayama
- Department of Thoracic Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Munetaka Masuda
- Department of Surgery, Yokohama City University, Yokohama, Japan
| |
Collapse
|
50
|
Grigoriadou GΙ, Esagian SM, Ryu HS, Nikas IP. Molecular Profiling of Malignant Pleural Effusions with Next Generation Sequencing (NGS): Evidence that Supports Its Role in Cancer Management. J Pers Med 2020; 10:jpm10040206. [PMID: 33139621 PMCID: PMC7712846 DOI: 10.3390/jpm10040206] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 12/15/2022] Open
Abstract
Malignant pleural effusions (MPEs) often develop in advanced cancer patients and confer significant morbidity and mortality. In this review, we evaluated whether molecular profiling of MPEs with next generation sequencing (NGS) could have a role in cancer management, focusing on lung cancer. We reviewed and compared the diagnostic performance of pleural fluid liquid biopsy with other types of samples. When applied in MPEs, NGS may have comparable performance with corresponding tissue biopsies, yield higher DNA amount, and detect more genetic aberrations than blood-derived liquid biopsies. NGS in MPEs may also be preferable to plasma liquid biopsy in advanced cancer patients with a MPE and a paucicellular or it could be difficult to obtain tissue/fine-needle aspiration biopsy. Of interest, post-centrifuge supernatant NGS may exhibit superior results compared to cell pellet, cell block or other materials. NGS in MPEs can also guide clinicians in tailoring established therapies and identifying therapy resistance. Evidence is still premature regarding the role of NGS in MPEs from patients with cancers other than lung. We concluded that MPE processing could provide useful prognostic and theranostic information, besides its diagnostic role.
Collapse
Affiliation(s)
- Georgia Ι. Grigoriadou
- 1st Department of Medical Oncology, Theageneio Anticancer Hospital, 54007 Thessaloniki, Greece;
| | - Stepan M. Esagian
- Department of Medicine, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Korea;
| | - Ilias P. Nikas
- School of Medicine, European University of Cyprus, 2404 Nicosia, Cyprus
- Correspondence:
| |
Collapse
|