1
|
Russo G, Scimone C, Palumbo L, Roscigno G, Sarracino C, Tomaiuolo I, Pisapia P, Pepe F, Rocco D, Gridelli C, Troncone G, Malapelle U. Biologics for novel driver altered non-small cell lung cancer: potential and pitfalls. Crit Rev Oncol Hematol 2025; 212:104748. [PMID: 40324663 DOI: 10.1016/j.critrevonc.2025.104748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025] Open
Abstract
Precision medicine has revolutionized clinical paradigm of lung cancer (LC) patients optimizing therapeutical options on the basis of molecular fingerprinting of tumor cells. The advent of the genomic era contributed to the widespread diffusion of sequencing technologies laying the basis for the approval of an increasing number of clinically relevant predictive biomarkers in clinical settings. In the rapidly evolving scenario of predictive biomarkers, mandatory testing genes demonstrated a statistically significant clinical benefit in LC patients elected to molecular tests, but emerging biomarkers are under investigation to raise the bar in the clinical management of LC patients. To date, promising IHC-based predictive biomarkers emerged as potentially integrative tools in the panel of clinically approved biomarkers. On this basis, genomic, transcriptomic and proteomic data are gaining ground toward "3D" biology" supporting the need of a multidimensional analysis of tumor cells to clinically stratify LC patients. Here we sought to overview the most promising biomarkers investigated in clinical trials to be integrated into diagnostic panel of predictive biomarkers tools for NSCLC patients.
Collapse
Affiliation(s)
- Gianluca Russo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Claudia Scimone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Lucia Palumbo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Giuseppina Roscigno
- Department of Biology, Complesso Universitario Monte Sant'Angelo, University of Naples Federico II, Via Cintia 4, 80126 Naples, Italy
| | - Claudia Sarracino
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Ilaria Tomaiuolo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Francesco Pepe
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Danilo Rocco
- Department of Pulmonary Oncology, AORN dei Colli Monaldi, Napoli, Italy
| | - Cesare Gridelli
- Division of Medical Oncology, 'S. G. Moscati' Hospital, Avellino, Italy
| | - Giancarlo Troncone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy.
| |
Collapse
|
2
|
Suriano I, Frasca L, Longo F, Sarubbi A, Tacchi G, Crucitti P. Diagnostic Yield of CE-EBUS in Mediastinal and Hilar Lymphadenopathy: A Preliminary Study. J Clin Med 2025; 14:2800. [PMID: 40283633 PMCID: PMC12027627 DOI: 10.3390/jcm14082800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/01/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Contrast-enhanced endobronchial ultrasound (CE-EBUS) is a minimally invasive technique that combines traditional endobronchial ultrasound (EBUS) with a contrast agent (sulfur hexafluoride), enhancing the visualization of blood flow in mediastinal and hilar lymph nodes. This study aimed to assess the use of CE-EBUS in patients with advanced neoplasms and hilar or mediastinal lymphadenopathy, particularly to improve diagnostic accuracy and expedite sample collection. Methods: A retrospective observational study was conducted from April 2021 to December 2023, involving 49 patients divided into two groups: EBUS (n = 26) and CE-EBUS (n = 23). Patients had advanced neoplasms with hilar and mediastinal lymphadenopathy, including bulky masses and nodal metastases with central necrosis. In the CE-EBUS group, 4.8 mL of sulfur hexafluoride was administered intravenously. Morphological, echogenic, and vascular characteristics, diagnostic accuracy, sample collection adequacy and molecular testing were compared between the groups. Results: The diagnostic accuracy in CE-EBUS was similar to EBUS (21 vs. 19 patients), with no significant difference (p = 0.100). However, for patients with bulky masses and necrosis, the molecular assessment rate was significantly higher in the CE-EBUS group (81.8%) compared to the EBUS group (33.3%) (p = 0.014). Conclusions: CE-EBUS-TBNA could improve the accuracy of molecular assessments in patients with bulky, necrotic lymphadenopathy and could help collect vital neoplastic tissue for molecular testing.
Collapse
Affiliation(s)
- Ilaria Suriano
- Department of Thoracic Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (F.L.); (A.S.); (G.T.); (P.C.)
| | - Luca Frasca
- Department of Thoracic Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (F.L.); (A.S.); (G.T.); (P.C.)
- PhD Course in Microbiology, Immunology, Infectious Diseases, and Transplants (MIMIT), University Tor Vergata, Viale Oxford, 81, 00133 Rome, Italy
| | - Filippo Longo
- Department of Thoracic Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (F.L.); (A.S.); (G.T.); (P.C.)
| | - Antonio Sarubbi
- Department of Thoracic Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (F.L.); (A.S.); (G.T.); (P.C.)
- Master’s Degree Program in Medicine and Surgery, Campus Bio-Medico University, Via Alvaro del Portillo, 21, 00128 Rome, Italy
| | - Giovanni Tacchi
- Department of Thoracic Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (F.L.); (A.S.); (G.T.); (P.C.)
| | - Pierfilippo Crucitti
- Department of Thoracic Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Rome, Italy; (F.L.); (A.S.); (G.T.); (P.C.)
| |
Collapse
|
3
|
Penault-Llorca F, Socinski MA. Emerging molecular testing paradigms in non-small cell lung cancer management-current perspectives and recommendations. Oncologist 2025; 30:oyae357. [PMID: 40126879 PMCID: PMC11966107 DOI: 10.1093/oncolo/oyae357] [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: 07/18/2024] [Accepted: 11/20/2024] [Indexed: 03/26/2025] Open
Abstract
Advances in molecular testing and precision oncology have transformed the clinical management of lung cancer, especially non-small cell lung cancer, enhancing diagnosis, treatment, and outcomes. Practical guidelines offer insights into selecting appropriate biomarkers and assays, emphasizing the importance of comprehensive testing. However, real-world data reveal the underutilization of biomarker testing and consequently targeted therapies. Molecular testing often occurs late in diagnosis or not at all in clinical practice, leading to delayed or inadequate treatment. Enhancing precision requires adherence to best practices by all health care professionals involved, which can ultimately improve lung cancer patient outcomes. The future of precision oncology for lung cancer will likely involve a more personalized approach, starting increasingly from earlier disease settings, with novel and more complex targeted therapies, immunotherapies, and combination regimens, and relying on liquid biopsies, muti-detection advanced genomic technologies and data integration, with artificial intelligence as a central orchestrator. This review presents the currently known actionable mutations in lung cancer and new upcoming ones that are likely to enter clinical practice soon and provides an overview of established and emerging concepts in testing methodologies. Challenges are discussed and best practice recommendations are made that are relevant today, will continue to be relevant in the future, and are likely to be relevant for other cancer types too.
Collapse
Affiliation(s)
- Frédérique Penault-Llorca
- Department of Pathology, Centre Jean Perrin, Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, Clermont Ferrand F-63000, France
| | - Mark A Socinski
- Oncology and Hematology, AdventHealth Cancer Institute, Orlando, FL 32804, United States
| |
Collapse
|
4
|
Wilson EM, Huang R, Jones KD, Hagemann IS, Temkin SM, McAlpine JN, Powell MA, Kepper MM, Hagemann AR. Challenges in implementation of molecular classification in early stage endometrial cancer-An NRG Oncology cooperative group mixed-methods study. Cancer 2025; 131:e35596. [PMID: 39420498 DOI: 10.1002/cncr.35596] [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: 06/30/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Professional guidelines recommend molecular profiling for mismatch repair (MMR), p53, and polymerase epsilon (POLE) status in endometrial cancer (EC). However, adoption in the United States has not been documented, and barriers to the implementation of testing have not been described. METHODS In this mixed-methods study, implementation science frameworks were used to develop a quantitative survey. Gynecologic oncologists, medical oncologists, radiation oncologists, and pathologists affiliated with NRG Oncology programs were contacted through snowball sampling and were surveyed during 2022-2023. A subset of respondents was interviewed. Statistical and thematic analyses were performed. RESULTS At least 403 NRG Oncology-affiliated providers were contacted for the survey, and 107 (26.6%) responded. Greater than 90% of respondents perceived POLE, MMR, and p53 status as important for clinical care. MMR and p53 tests were perceived as easy to obtain, but only 24.2% of respondents reported that POLE testing was moderately or very easy to obtain. Respondents from academic sites reported better access to molecular classification and perceived greater importance of molecular classification compared with respondents from community sites. In thematic analysis of 13 qualitative interviews, cost concerns were reported as large barriers to testing. Interviewees reported a desire for prospective data to guide treatment selection based on classification results. CONCLUSIONS Although integrating molecular classification into standard pathologic reporting is recommended, and clinicians perceive molecular profiling in early stage EC as important, survey respondents noted significant implementation barriers. Implementation challenges that differ between community oncology and academic practice settings were identified. Strategies to improve equitable access to molecular classification of early stage EC are needed.
Collapse
Affiliation(s)
- Elise M Wilson
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University in St Louis, St Louis, Missouri, USA
- Division of Gynecologic Oncology, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, San Diego, California, USA
| | - Ruizhi Huang
- The Advanced Health Data (AHEAD) Institute, St Louis University School of Medicine, St Louis, Missouri, USA
| | - Kristen D Jones
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University in St Louis, St Louis, Missouri, USA
| | - Ian S Hagemann
- Department of Pathology and Immunology, Washington University in St Louis, St Louis, Missouri, USA
| | - Sarah M Temkin
- National Institutes of Health Office of Research on Womens Health, Bethesda, Maryland, USA
| | - Jessica N McAlpine
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Matthew A Powell
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University in St Louis, St Louis, Missouri, USA
| | - Maura M Kepper
- George W. Brown School of Social Work, Washington University in St Louis, St Louis, Missouri, USA
| | - Andrea R Hagemann
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University in St Louis, St Louis, Missouri, USA
| |
Collapse
|
5
|
Roy‐Chowdhuri S, Mani H, Fox AH, Tsao A, Sholl LM, Farjah F, Johnson BE, Osarogiagbon RU, Rivera MP, Silvestri GA, Smith RA, Wistuba II. The American Cancer Society National Lung Cancer Roundtable strategic plan: Methods for improving turnaround time of comprehensive biomarker testing in non-small cell lung cancer. Cancer 2024; 130:4200-4212. [PMID: 39347608 PMCID: PMC11585344 DOI: 10.1002/cncr.34926] [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: 09/14/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 10/01/2024]
Abstract
Comprehensive biomarker testing for patients with non-small cell lung cancer is critical for selecting appropriate targeted therapy or immunotherapy. Ensuring timely ordering, processing, and reporting is key to optimizing patient outcomes. However, various factors can prevent or delay patients from being offered the option of treatment selection based on comprehensive biomarker testing. These factors include problems with access to testing, tissue adequacy, turnaround time, and health insurance coverage and billing practices. Turnaround time depends on several logistical and tissue handling factors, which involve institutional policies, processes, resources, testing methodology, and testing algorithms that vary across different practices. In this article, the authors identify key factors that prolong biomarker testing turnaround time, propose strategies to reduce it, and present a process map to aid physicians and key organizational stakeholders in improving testing efficiency.
Collapse
Affiliation(s)
- Sinchita Roy‐Chowdhuri
- Division of Pathology and Laboratory MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Haresh Mani
- Department of PathologyInova Fairfax HospitalFalls ChurchVirginiaUSA
| | - Adam H. Fox
- Division of Pulmonary Critical CareAllergy and Sleep MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Anne Tsao
- Department of Thoracic/Head and Neck Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Lynette M. Sholl
- Brigham and Women's Hospital and Department of PathologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Farhood Farjah
- Department of SurgeryUniversity of WashingtonSeattleWashingtonUSA
| | - Bruce E. Johnson
- Department of Medical OncologyDana‐Farber Cancer InstituteHarvard Medical SchoolBostonMassachusettsUSA
| | | | - M. Patricia Rivera
- Department of Medicine, Division of Pulmonary and Critical Care MedicineWilmot Cancer Institute, The University of Rochester Medical CenterRochesterNew YorkUSA
| | - Gerard A. Silvestri
- Division of Pulmonary Critical CareAllergy and Sleep MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Robert A. Smith
- Center for Early Cancer Detection ScienceAmerican Cancer SocietyAtlantaGeorgiaUSA
| | - Ignacio I. Wistuba
- Division of Pathology and Laboratory MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| |
Collapse
|
6
|
Bestvina CM, Waters D, Morrison L, Emond B, Lafeuille MH, Hilts A, Mujwara D, Lefebvre P, He A, Vanderpoel J. Impact of next-generation sequencing vs polymerase chain reaction testing on payer costs and clinical outcomes throughout the treatment journeys of patients with metastatic non-small cell lung cancer. J Manag Care Spec Pharm 2024; 30:1467-1478. [PMID: 39259000 DOI: 10.18553/jmcp.2024.24137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
BACKGROUND For patients with metastatic non-small cell lung cancer (mNSCLC), next-generation sequencing (NGS) biomarker testing has been associated with a faster time to appropriate targeted therapy and more comprehensive testing relative to polymerase chain reaction (PCR) testing. However, the impact on payer costs and clinical outcomes during patients' treatment journeys has not been fully characterized. OBJECTIVE To assess the costs and clinical outcomes of NGS vs PCR biomarker testing among patients with newly diagnosed de novo mNSCLC from a US payers' perspective. METHODS A Markov model assessed costs and clinical outcomes of NGS vs PCR testing from the start of testing up to 3 years after. Patients entered the model after receiving biomarker test results and then initiated first-line (1L) targeted or nontargeted therapy (immunotherapy and/or chemotherapy) depending on actionable mutation detection. A few patients with an actionable mutation were not detected by PCR and inappropriately initiated 1L nontargeted therapy. At each 1-month cycle, patients could remain on treatment with 1L, progress to second line or later (2L+), or die. Literature-based inputs included the rates of progression-free survival (PFS) and overall survival (OS), targeted and nontargeted therapy costs, total costs of testing, and medical costs of 1L, 2L+, and death. Per patient average PFS and OS as well as cumulative costs were reported for NGS and PCR testing. RESULTS In a modeled population of 100 patients (75% commercial and 25% Medicare), 45.9% of NGS and 40.0% of PCR patients tested positive for an actionable mutation. Relative to PCR, NGS was associated with $7,386 in savings per patient (NGS = $326,154; PCR = $333,540) at 1 year, driven by lower costs of testing, including estimated costs of delayed care and nontargeted therapy initiation before receiving test results (NGS = $8,866; PCR = $16,373). Treatment costs were similar (NGS = $305,644; PCR = $305,283). In the PCR cohort, the per patient costs of inappropriate 1L nontargeted therapy owing to undetected mutations were $6,455, $6,566, and $6,569 over the first 1, 2, and 3 years, respectively. Relative to PCR testing, NGS was associated with $4,060 in savings at 2 years and $1,092 at 3 years. Patients who initiated 1L targeted therapy had an additional 5.4, 8.8, and 10.4 months of PFS and an additional 1.4, 3.6, and 5.3 months of OS over the first 1, 2, and 3 years, respectively, relative to those who inappropriately initiated 1L nontargeted therapy. CONCLUSIONS In this Markov model, as early as year 1, and over 3 years following biomarker testing, patients with newly diagnosed de novo mNSCLC undergoing NGS testing are projected to have cost savings and longer PFS and OS relative to those tested with PCR.
Collapse
Affiliation(s)
| | - Dexter Waters
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA
| | | | | | | | | | | | | | - Andy He
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA
| | - Julie Vanderpoel
- Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA
| |
Collapse
|
7
|
Fox AH, Alexander M, Forcucci JA, Silvestri GA. Biomarker Testing for Guiding Precision Medicine for Patients With Non-Small Cell Lung Cancer. Chest 2024; 166:1239-1249. [PMID: 39151823 DOI: 10.1016/j.chest.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024] Open
Abstract
The initial management of patients with lung cancer is growing more complex in the context of an expanding number of precision medicine treatments. These challenges are accompanied by opportunities to deliver more efficacious and less toxic treatments to patients. Indications for these treatments are also expanding, and patients with lung cancer across multiple stages now require biomarker testing. Given their role in the initial management of patients being diagnosed with lung cancer, pulmonologists must have fundamental knowledge regarding the importance, indications, and implications of biomarker testing across the spectrum of histology and stage. The purpose of this review is to provide fundamental knowledge regarding biomarker testing, its incorporation into the initial diagnostic and staging evaluation, and guidance for working within a multidisciplinary team to achieve timely and comprehensive biomarker testing to direct the use of precision medicine treatments.
Collapse
Affiliation(s)
- Adam H Fox
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Medical University of South Carolina, Charleston, SC.
| | - Mariam Alexander
- Division of Hematology and Oncology, Medical University of South Carolina, Charleston, SC
| | - Jessica A Forcucci
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC
| | - Gerard A Silvestri
- Division of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
8
|
Caputo A, Pisapia P, L'Imperio V. Current role of cytopathology in the molecular and computational era: The perspective of young pathologists. Cancer Cytopathol 2024; 132:678-685. [PMID: 38748507 DOI: 10.1002/cncy.22832] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 11/03/2024]
Abstract
Cytopathology represents a well established diagnostic approach because of its limited cost, reliability, and minimal invasiveness with respect to other methodologies. The evolving complexity of the different classifications systems and the implementation of ancillary techniques to refine the diagnosis is progressively helping in the risk of malignancy stratification, and the adoption of next-generation sequencing techniques contributes to enrich this valuable tool with predictive information, which is always more essential in the tailored medicine era. The recent introduction of digital and computational pathology is further boosting the potentialities of cytopathology, aiding in the interpretation of samples to improve the cost effectiveness of large screening programs and the diagnostic efficiency within intermediate/atypical categories. Moreover, the adoption of artificial intelligence tools is promising to complement molecular investigations, representing a stimulating perspective in the cytopathology field. In this work, the authors tried to summarize the multifaceted nature of this complex and evolving field of pathology, synthesizing the most recent advances and providing the young pathologists' perspective on this fascinating world.
Collapse
Affiliation(s)
- Alessandro Caputo
- Department of Medicine and Surgery, University of Salerno, Fisciano, Italy
| | - Pasquale Pisapia
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Vincenzo L'Imperio
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo dei Tintori, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
9
|
Al Mana AF, Culp K, Keeler A, Perrera O, Rajagopalan M, Jacky L, Brown B, Thyagarajan B. Performance of a Rapid Digital PCR Test for the Detection of Non-Small Cell Lung Cancer (NSCLC) Variants. Mol Diagn Ther 2024; 28:791-802. [PMID: 39093547 DOI: 10.1007/s40291-024-00732-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Next-generation sequencing is widely used for comprehensive molecular profiling for many cancers including lung cancer. However, the complex workflows and long turnaround times limit its access and utility. ChromaCode's High Definition PCR Non-Small Cell Lung Cancer Panel (HDPCR™ NSCLC Panel) is a low-cost, rapid turnaround, digital polymerase chain reaction assay that is designed to detect variants in nine NSCLC genes listed in National Comprehensive Cancer Network guidelines. METHODS This assay uses TaqMan® probe limiting chemistry and proprietary analysis software to enable multi-target detection within a single-color channel. We compared the performance of the HDPCR™ NSCLC Panel against an in-house, laboratory-developed, targeted next-generation sequencing panel used in the Molecular Diagnostics Laboratory at the University of Minnesota Medical Center to detect biomarkers for NSCLC. RESULTS The overall accuracy of the HDPCR panel was 99.48% (95% confidence interval 99.01-99.76) with a sensitivity of 95.35% (95% confidence interval 88.52-98.72) and a specificity of 99.69% (95% confidence interval 99.29-99.90). The HDPCR wet lab workflow was 4 h, and the time to generate variant calls from raw data using the ChromaCode Cloud was 2 minutes. CONCLUSIONS We demonstrated that the HDPCR™ NSCLC Panel provides timely, comprehensive, and sensitive mutation detection in NSCLC samples with results in less than 24 h.
Collapse
Affiliation(s)
- Abdulaziz F Al Mana
- Department of Laboratory Medicine and Pathology, University of Minnesota, 100 Church St SE, Minneapolis, MN, 55455, USA
| | | | - Abby Keeler
- Department of Laboratory Medicine and Pathology, University of Minnesota, 100 Church St SE, Minneapolis, MN, 55455, USA
| | | | | | | | | | - Bharat Thyagarajan
- Department of Laboratory Medicine and Pathology, University of Minnesota, 100 Church St SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
10
|
Kowalski DM, Zaborowska-Szmit M, Bryl M, Byszek A, Dziedzic DA, Jaśkiewicz P, Langfort R, Krzakowski M, Orłowski T, Ramlau R, Szmit S. The Detailed Analysis of Polish Patients with Non-Small Cell Lung Cancer Through Insights from Molecular Testing (POL-MOL Study). Int J Mol Sci 2024; 25:11354. [PMID: 39518907 PMCID: PMC11547071 DOI: 10.3390/ijms252111354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Molecular testing is recommended in patients with metastatic non-small cell lung cancer (NSCLC), but the extent of its use in Poland is unknown. The aim of the POL-MOL study was to investigate the frequency of using molecular testing in Polish patients with NSCLC. The invited Polish oncologists completed two questionnaires, and data for 1001 patients undergoing systemic treatment for NSCLC were collected. The use of molecular tests for the following genetic mutations was recorded: EGFR (del19, sub21), EGFR (other than del19/sub21), EGFR T790M, ALK (expression and rearrangement), RET, NTRK, ROS1, BRAF, HER2, and MET, as well as for immunochemical assessment of programmed cell death ligand 1 (PD-L1). Thanks to the weighting procedure, the results are representative of the population of Polish patients treated for NSCLC. Molecular tests were applied in 78% of patients with NSCL, 70% of patients with NSCLC not otherwise specified, and in 12% of patients with squamous cell carcinoma of the lung. The frequency of application increased with disease stage in all groups. In patients with squamous cell carcinoma, approximately 30% of tests for EGFR, ALK, and RET mutations were positive, which confirms the importance of testing at least a preselected subgroup of patients.
Collapse
Affiliation(s)
- Dariusz M. Kowalski
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (D.M.K.); (M.Z.-S.); (P.J.); (M.K.)
- Polish Lung Cancer Study Group, 01-138 Warsaw, Poland (A.B.); (D.A.D.); (R.L.); (T.O.); (R.R.)
| | - Magdalena Zaborowska-Szmit
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (D.M.K.); (M.Z.-S.); (P.J.); (M.K.)
- Polish Lung Cancer Study Group, 01-138 Warsaw, Poland (A.B.); (D.A.D.); (R.L.); (T.O.); (R.R.)
| | - Maciej Bryl
- Polish Lung Cancer Study Group, 01-138 Warsaw, Poland (A.B.); (D.A.D.); (R.L.); (T.O.); (R.R.)
- Centre of Pulmonology and Thoracic Surgery, 60-569 Poznań, Poland
| | - Agnieszka Byszek
- Polish Lung Cancer Study Group, 01-138 Warsaw, Poland (A.B.); (D.A.D.); (R.L.); (T.O.); (R.R.)
| | - Dariusz Adam Dziedzic
- Polish Lung Cancer Study Group, 01-138 Warsaw, Poland (A.B.); (D.A.D.); (R.L.); (T.O.); (R.R.)
- Department of Thoracic Surgery, National Institute of Tuberculosis and Pulmonary Disease, 01-138 Warsaw, Poland
| | - Piotr Jaśkiewicz
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (D.M.K.); (M.Z.-S.); (P.J.); (M.K.)
- Polish Lung Cancer Study Group, 01-138 Warsaw, Poland (A.B.); (D.A.D.); (R.L.); (T.O.); (R.R.)
| | - Renata Langfort
- Polish Lung Cancer Study Group, 01-138 Warsaw, Poland (A.B.); (D.A.D.); (R.L.); (T.O.); (R.R.)
- Department of Pathology, National Institute of Tuberculosis and Pulmonary Disease, 01-138 Warsaw, Poland
| | - Maciej Krzakowski
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (D.M.K.); (M.Z.-S.); (P.J.); (M.K.)
- Polish Lung Cancer Study Group, 01-138 Warsaw, Poland (A.B.); (D.A.D.); (R.L.); (T.O.); (R.R.)
| | - Tadeusz Orłowski
- Polish Lung Cancer Study Group, 01-138 Warsaw, Poland (A.B.); (D.A.D.); (R.L.); (T.O.); (R.R.)
- Department of Thoracic Surgery, National Institute of Tuberculosis and Pulmonary Disease, 01-138 Warsaw, Poland
| | - Rodryg Ramlau
- Polish Lung Cancer Study Group, 01-138 Warsaw, Poland (A.B.); (D.A.D.); (R.L.); (T.O.); (R.R.)
- Institute of Oncology, Poznan University of Medical Sciences, 60-514 Poznań, Poland
| | - Sebastian Szmit
- Polish Lung Cancer Study Group, 01-138 Warsaw, Poland (A.B.); (D.A.D.); (R.L.); (T.O.); (R.R.)
- Department of Cardio-Oncology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
- Department of Cancer Diagnostics and Cardio-Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| |
Collapse
|
11
|
Gristina V, Russo TDB, Barraco N, Gottardo A, Pepe F, Russo G, Fulfaro F, Incorvaia L, Badalamenti G, Troncone G, Malapelle U, Russo A, Bazan V, Galvano A. Clinical utility of ctDNA by amplicon based next generation sequencing in first line non small cell lung cancer patients. Sci Rep 2024; 14:22141. [PMID: 39333636 PMCID: PMC11436775 DOI: 10.1038/s41598-024-73046-y] [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: 05/29/2024] [Accepted: 09/12/2024] [Indexed: 09/29/2024] Open
Abstract
The assessment of ctDNA has emerged as a minimally invasive avenue for molecular diagnosis and real-time tracking of tumor progression in NSCLC. However, the evaluation of ctDNA by amplicon-based NGS has been not endorsed by all the healthcare systems and remains to be fully integrated into clinical routine practice. To compare tissue single-gene with plasma multiplexed testing, we retrospectively evaluated 120 plasma samples from 12 consecutive patients with advanced non-squamous NSCLC who were part of a prospective study enrolling treatment-naïve patients and in which tissue samples were evaluated using a single-gene testing approach. While the plasma ctDNA detection of EGFR and BRAF mutations had an acceptable level of concordance with the archival tissue (85%), discordance was seen in all the patients in whom ALK alterations were only detected in tissue samples. Among six responders and six non-responders, early ctDNA mutant allelic frequency (MAF) reduction seemed to predict radiologic responses and longer survival, whereas increasing MAF values with the emergence of co-mutations like BRAFV600E, KRASG12V or TP53M237I seemed to be an early indicator of molecular and radiologic progression. This report using an amplicon-based NGS assay on ctDNA underscores the real-life need for plasma and tissue genotyping as complementary tools in the diagnostic and therapeutic decision-making process.
Collapse
Affiliation(s)
- Valerio Gristina
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Tancredi Didier Bazan Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Nadia Barraco
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Andrea Gottardo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Gianluca Russo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Fabio Fulfaro
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Lorena Incorvaia
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy.
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Antonio Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy.
| | - Viviana Bazan
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Palermo, Italy
| | - Antonio Galvano
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| |
Collapse
|
12
|
Sposito M, Eccher S, Pasqualin L, Scaglione IM, Avancini A, Tregnago D, Trestini I, Insolda J, Bonato A, Ugel S, Derosa L, Milella M, Pilotto S, Belluomini L. Characterizing the immune tumor microenvironment in ALK fusion-positive lung cancer: state-of-the-art and therapeutical implications. Expert Rev Clin Immunol 2024; 20:959-970. [PMID: 38913940 DOI: 10.1080/1744666x.2024.2372327] [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: 03/11/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
INTRODUCTION Approximately 5% of non-small cell lung cancer (NSCLC), exhibits anaplastic lymphoma kinase (ALK) rearrangements. EML4-ALK fusions account for over 90% of ALK rearrangements in NSCLC. The advent of treatment targeting ALK has significantly improved survival rates in patients with advanced ALK-positive NSCLC. However, the emergence of resistance mechanisms and the subsequent progression disease inevitably occurs. The tumor immune microenvironment (TIME) plays a pivotal role in lung cancer, influencing disease development, patient's outcomes, and response to treatments. AREAS COVERED The aim of this review is to provide a comprehensive characterization of the TIME in ALK rearranged NSCLC and its intrinsic plasticity under treatment pressure. EXPERT OPINION Recognizing the fundamental role of the TIME in cancer progression has shifted the paradigm from a tumor cell-centric perspective to the understanding of a complex tumor ecosystem. Understanding the intricate dynamics of the TIME, its influence on treatment response, and the potential of immunotherapy in patients with ALK-positive NSCLC are currently among the primary research objectives in this patient population.
Collapse
Affiliation(s)
- Marco Sposito
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Serena Eccher
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Luca Pasqualin
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Ilaria Mariangela Scaglione
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Alice Avancini
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Daniela Tregnago
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Ilaria Trestini
- Dietetic Service, Hospital Medical Direction, University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - Jessica Insolda
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Adele Bonato
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Santa Chiara Hospital, Pisa, Italy
| | - Stefano Ugel
- Immunology Section, University Hospital and Department of Medicine, University of Verona, Verona, Italy
| | - Lisa Derosa
- INSERM U1015 Gustave Roussy Cancer Campus, Villejuif Cedex, Villejuif, France
- Faculté de Médicine, Université Paris-Saclay, Le Kremlin-Bicetre, France
| | - Michele Milella
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Sara Pilotto
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Lorenzo Belluomini
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| |
Collapse
|
13
|
Shang J, Kondowe B, Zhang H, Xie X. Identifying ribonucleotide reductase subunit genes as potential lung adenocarcinomas biomarkers using integrated bioinformatics analysis. Malawi Med J 2024; 36:134-143. [PMID: 40191558 PMCID: PMC11970203 DOI: 10.4314/mmj.v36i2.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
Introduction Dysregulation of ribonucleotide reductase (RR) subunit genes (RRM1, RRM2 and RRM2B) expression is reported to be involved in the occurrence of various human malignancies. However, the prognostic value of RR subunit genes expression in lung adenocarcinoma (LUAD) patients remains controversial. Objective This study aims to analyze the expression profiles, prognostic values, and immune infiltrating associations of RR subunit genes in LUAD to explore whether RR subunit gene expression has value in the prognosis of patients with lung adenocarcinoma (LUAD). Methodology We used multiple search engines to access multiple online bioinformatics databases, including Oncomine, TIMER, GEPIA, Kaplan-Meier Plotter, PrognoScan, the Human Protein Atlas, MD Anderson Cancer Center, UCSC Xena, cBioProtal, TCGA, GEO, DAVID, and STRING databases. Results The study found that RRM1 and RRM2 might be an attractive target for treating LUAD, while RRM2B were down-expressed in LUAD (P < 0.05). The study also found that high RRM1 or RRM2 expression, or low RRM2B expression suggested poor prognosis of LUAD patients in both TCGA and GEO databases (P < 0.05). Additionally, our results indicated that RR subunit genes expressions have different characteristics with immune infiltrating, RRM2B had a slight but significant positive correlation with almost every infiltrating immune cells except CD4+ T cells (all P < 0.05). Furthermore, by co-expression gene network analysis of RR subunit genes, we found that five new hub genes (PLK1, AURKA, CDCA8, TTK and CDC45) were significantly positively correlated with RRM1 and RRM2 expression whereas were negatively correlated with RRM2B expression, and these five hub genes were identified to be related with a poor prognosis in LUAD patients (P < 0.05). Conclusion The study findings demonstrate that RR subunit genes may be a prognostic marker and therapeutic target for LUAD patients.
Collapse
Affiliation(s)
- Jin Shang
- Department of Medical Imaging, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | | | - Hui Zhang
- Department of Medical Imaging, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Xinming Xie
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| |
Collapse
|
14
|
Malapelle U, Leighl N, Addeo A, Hershkovitz D, Hochmair MJ, Khorshid O, Länger F, de Marinis F, Peled N, Sheffield BS, Smit EF, Viteri S, Wolf J, Venturini F, O'Hara RM, Rolfo C. Recommendations for reporting tissue and circulating tumour (ct)DNA next-generation sequencing results in non-small cell lung cancer. Br J Cancer 2024; 131:212-219. [PMID: 38750115 PMCID: PMC11263606 DOI: 10.1038/s41416-024-02709-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/25/2024] [Accepted: 04/30/2024] [Indexed: 07/24/2024] Open
Abstract
Non-small cell lung cancer is a heterogeneous disease and molecular characterisation plays an important role in its clinical management. Next-generation sequencing-based panel testing enables many molecular alterations to be interrogated simultaneously, allowing for comprehensive identification of actionable oncogenic drivers (and co-mutations) and appropriate matching of patients with targeted therapies. Despite consensus in international guidelines on the importance of broad molecular profiling, adoption of next-generation sequencing varies globally. One of the barriers to its successful implementation is a lack of accepted standards and guidelines specifically for the reporting and clinical annotation of next-generation sequencing results. Based on roundtable discussions between pathologists and oncologists, we provide best practice recommendations for the reporting of next-generation sequencing results in non-small cell lung cancer to facilitate its use and enable easy interpretation for physicians. These are intended to complement existing guidelines related to the use of next-generation sequencing (solid and liquid). Here, we discuss next-generation sequencing workflows, the structure of next-generation sequencing reports, and our recommendations for best practice thereof. The aim of these recommendations and considerations is ultimately to ensure that reports are fully interpretable, and that the most appropriate treatment options are selected based on robust molecular profiles in well-defined reports.
Collapse
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Natasha Leighl
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Alfredo Addeo
- Oncology Unit, Geneva University Hospital, Geneva, Switzerland
| | | | - Maximilian J Hochmair
- Department of Respiratory & Critical Care Medicine, Karl Landsteiner Institute of Lung Research & Pulmonary Oncology, Klinik Floridsdorf, Vienna, Austria
| | - Ola Khorshid
- National Cancer Institute, Cairo University, Cairo, Egypt
| | - Florian Länger
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Filippo de Marinis
- Division of Thoracic Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Nir Peled
- Helmesely Cancer Center, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Brandon S Sheffield
- Division of Advanced Diagnostics, William Osler Health System, Brampton, ON, Canada
| | - Egbert F Smit
- Department of Pulmonary Diseases, Leiden University Medical Centre, Leiden, The Netherlands
| | - Santiago Viteri
- UOMI Cancer Center, Clínica Mi Tres Torres, Barcelona, Spain
| | - Jürgen Wolf
- Lung Cancer Group Cologne, Center for Integrated Oncology, University Hospital of Cologne, Cologne, Germany
| | | | | | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, New York, NY, USA.
| |
Collapse
|
15
|
Hirsch FR, Kim C. The Importance of Biomarker Testing in the Treatment of Advanced Non-Small Cell Lung Cancer: A Podcast. Oncol Ther 2024; 12:223-231. [PMID: 38536631 PMCID: PMC11187040 DOI: 10.1007/s40487-024-00271-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 03/06/2024] [Indexed: 06/20/2024] Open
Abstract
The identification of actionable biomarkers and development of targeted therapies have revolutionized the field of lung cancer treatment. In patients with advanced non-small cell lung cancer (NSCLC), biomarker testing can inform selection of effective targeted therapies as well as avoid therapies that are less likely to be effective in certain populations. A growing number of actionable targets, including those involving EGFR, ALK, ROS1, BRAF, MET, KRAS, NTRK, RET, HER2, and PD-L1, can be identified with biomarker testing. More than half of patients with advanced NSCLC have tumors that harbor genetic alterations that can be targeted. When these patients are treated with targeted therapy, survival and quality of life may be significantly improved. In addition, broad-based molecular testing may detect alterations identifying patients who are potentially eligible for current or future clinical trials. Comprehensive biomarker testing rates in communities are often low, and turnaround times for results can be unacceptably long. There is an unmet need for widespread, efficient, and routine testing of all biomarkers recommended by clinical guidelines. New testing techniques and technologies can make this an attainable goal. Panel-based sequencing platforms are becoming more accessible, and molecular biomarker analysis of circulating tumor DNA is becoming more common. In this podcast, we discuss the importance of biomarker testing in advanced NSCLC and explore topics such as testing methodologies, effect of biomarker testing on patient outcomes, emerging technologies, and strategies for improving testing rates in the United States. Supplementary file1 (MP4 121301 KB).
Collapse
Affiliation(s)
- Fred R Hirsch
- Icahn School of Medicine, Center for Thoracic Oncology, Tisch Cancer Center, Mount Sinai, New York, NY, USA.
| | - Chul Kim
- Georgetown University, Washington, DC, USA
| |
Collapse
|
16
|
Shafiq I, Isse S, Khan N, Uzebeck M, Zoumot Z, Shabeer S, Wahla A. A retrospective, descriptive analysis identifying non‑small cell lung cancer molecular markers. Mol Clin Oncol 2024; 20:41. [PMID: 38756870 PMCID: PMC11097133 DOI: 10.3892/mco.2024.2738] [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: 11/13/2023] [Accepted: 03/27/2024] [Indexed: 05/18/2024] Open
Abstract
Non-small-cell lung cancer (NSCLC) remains one of the leading causes of cancer mortality worldwide. The aim of the present study was to review the histologic patterns and molecular drivers of NSCLC in patients with lung cancer. The electronic health records (EHR) of all patients diagnosed with lung cancer between April 2015 and September 2022 were obtained from a tertiary care hospital and retrospectively analysed. A total of 224 patients were identified of which 192 (138 males and 54 females) were included in the final analysis. Adenocarcinoma was the most common type of lung cancer identified, and accounted for 134 patients (70%), followed by squamous cell carcinoma in 47 (24%) patients, while large cell lung cancer was noted in only 5 (3%) patients. The most common mutations were EGFR mutations and were detected in 29 (15%) patients, followed by PD-L1 expression which was present in 56 (24.7%) patients, KRAS in 16 (8.3%) patients, ALK1 in 8 (4.2%) patients and BRAF, ROS1 and MET were present in 3 (1.6%), 2 (1%) and 1 (0.5%), respectively. The findings from the present study offer important insights into the epidemiological, clinical and molecular characteristics of NSCLC. Further research is warranted to explore the clinical implications of these findings.
Collapse
Affiliation(s)
- Irfan Shafiq
- Respiratory Institute, Cleveland Clinic Abu Dhabi, Al Maryah Island, Abu Dhabi 112412, United Arab Emirates
| | - Said Isse
- Respiratory Institute, Cleveland Clinic Abu Dhabi, Al Maryah Island, Abu Dhabi 112412, United Arab Emirates
| | - Naureen Khan
- Respiratory Institute, Cleveland Clinic Abu Dhabi, Al Maryah Island, Abu Dhabi 112412, United Arab Emirates
| | - Mateen Uzebeck
- Respiratory Institute, Cleveland Clinic Abu Dhabi, Al Maryah Island, Abu Dhabi 112412, United Arab Emirates
| | - Zaid Zoumot
- Respiratory Institute, Cleveland Clinic Abu Dhabi, Al Maryah Island, Abu Dhabi 112412, United Arab Emirates
| | - Safia Shabeer
- Respiratory Institute, Cleveland Clinic Abu Dhabi, Al Maryah Island, Abu Dhabi 112412, United Arab Emirates
| | - Ali Wahla
- Respiratory Institute, Cleveland Clinic Abu Dhabi, Al Maryah Island, Abu Dhabi 112412, United Arab Emirates
| |
Collapse
|
17
|
Cafaro A, Foca F, Nanni O, Chiumente M, Coppola M, Russi A, Svegliati E, Baldo P, Orzetti S, Enrico F, Foglio F, Pinnavaia D, Ladisa V, Lauria Pantano C, Lerose R, Nardulli P, Ferraiuolo S, Maiolino P, De Stasio I, Gradellini F, Gasbarro AR, Santeramo R, Carrucciu G, Provasi R, Cirino M, Cappelletto PC, Fonzi E, Pasqualini A, Vecchia S, Veraldi M, De Francesco AE, Crinò L, Delmonte A, Masini C. Real-World Safety and Outcome of First-Line Pembrolizumab Monotherapy for Metastatic NSCLC with PDL-1 Expression ≥ 50%: A National Italian Multicentric Cohort (" PEMBROREAL" Study). Cancers (Basel) 2024; 16:1802. [PMID: 38791882 PMCID: PMC11119961 DOI: 10.3390/cancers16101802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Results from the phase III Keynote-024 clinical trial established pembrolizumab monotherapy as the first-line standard of care for patients with metastatic NSCLC who have PD-L1 expression ≥ 50%, EGFR, and ALK wild-type tumors. However, given the differences between patients treated in routine clinical practice and those treated in a clinical trial, real-world data are needed to confirm the treatment benefit in standard practice. Given the lack of data on large cohorts of patients with long follow-ups, we designed an observational retrospective study of patients with metastatic NSCLC who were treated with pembrolizumab, starting from its reimbursement eligibility until December 2020. The primary endpoints were PFS and OS, determined using the Kaplan-Meier method. Response and safety were also evaluated. We followed 880 patients (median follow-up: 35.1 months) until February 2022. Median PFS and OS were 8.6 months (95% CI: 7.6-10.0) and 25.5 months (95% CI: 21.8-31.6), respectively. We also found that ECOG PS, PD-L1 expression, and habitual smoking were prognostic factors for PFS, while age, sex, ECOG PS, habitual smoking and histology had an impact on OS. Multivariable analysis confirms the prognostic role of PD-L1 for PFS and of ECOG for both PFS and OS. 39.9% of patients reported an adverse event, but only 6.3% of patients discontinued therapy due to toxicity. Our results suggest a long-term benefit of pembrolizumab in the first-line setting, as well as a safety profile consistent with the results of Keynote-024. Many collected variables appear to influence clinical outcome, but results from these exploratory unadjusted analyses should be interpreted with caution.
Collapse
Affiliation(s)
- Alessandro Cafaro
- Pharmacy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Flavia Foca
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (F.F.); (O.N.)
| | - Oriana Nanni
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (F.F.); (O.N.)
| | - Marco Chiumente
- Scientific Direction, Società Italiana di Farmacia Clinica e Terapia (SIFaCT), 10123 Turin, Italy;
| | - Marina Coppola
- Pharmacy Unit, IRCCS Istituto Oncologico Veneto (IOV), 35128 Padova, Italy; (M.C.); (A.R.); (E.S.)
| | - Alberto Russi
- Pharmacy Unit, IRCCS Istituto Oncologico Veneto (IOV), 35128 Padova, Italy; (M.C.); (A.R.); (E.S.)
| | - Elena Svegliati
- Pharmacy Unit, IRCCS Istituto Oncologico Veneto (IOV), 35128 Padova, Italy; (M.C.); (A.R.); (E.S.)
| | - Paolo Baldo
- Pharmacy Unit, CRO Aviano IRCCS, National Cancer Institute, 33081 Aviano, Italy; (P.B.); (S.O.)
| | - Sabrina Orzetti
- Pharmacy Unit, CRO Aviano IRCCS, National Cancer Institute, 33081 Aviano, Italy; (P.B.); (S.O.)
| | - Fiorenza Enrico
- Hospital Pharmacy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy; (F.E.); (F.F.); (D.P.)
| | - Federico Foglio
- Hospital Pharmacy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy; (F.E.); (F.F.); (D.P.)
| | - Davide Pinnavaia
- Hospital Pharmacy, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, 10060 Turin, Italy; (F.E.); (F.F.); (D.P.)
| | - Vito Ladisa
- Hospital Pharmacy, IRCCS National Cancer Institute Foundation, 20133 Milan, Italy; (V.L.); (C.L.P.)
| | - Claudia Lauria Pantano
- Hospital Pharmacy, IRCCS National Cancer Institute Foundation, 20133 Milan, Italy; (V.L.); (C.L.P.)
| | - Rosa Lerose
- Hospital Pharmacy, IRCCS-CROB Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy;
| | - Patrizia Nardulli
- Pharmacy Unit, National Cancer Research Center Istituto Tumori “Giovanni Paolo II”, 70121 Bari, Italy; (P.N.); (S.F.)
| | - Simona Ferraiuolo
- Pharmacy Unit, National Cancer Research Center Istituto Tumori “Giovanni Paolo II”, 70121 Bari, Italy; (P.N.); (S.F.)
| | - Piera Maiolino
- Pharmacy Unit, Istituto Nazionale Tumori “Fondazione G. Pascale”, IRCCS, 80131 Naples, Italy; (P.M.); (I.D.S.)
| | - Immacolata De Stasio
- Pharmacy Unit, Istituto Nazionale Tumori “Fondazione G. Pascale”, IRCCS, 80131 Naples, Italy; (P.M.); (I.D.S.)
| | - Federica Gradellini
- Pharmacy Unit, Azienda USL-IRCCS di Reggio Emilia, 42122 Reggio Emilia, Italy;
| | - Anna Rita Gasbarro
- Pharmacy Unit, University Hospital Policlinico, 70100 Bari, Italy; (A.R.G.); (R.S.)
| | - Rossella Santeramo
- Pharmacy Unit, University Hospital Policlinico, 70100 Bari, Italy; (A.R.G.); (R.S.)
| | | | - Riccardo Provasi
- Department of Pharmacy, Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.P.); (M.C.)
| | - Mario Cirino
- Department of Pharmacy, Institute for Maternal and Child Health-IRCCS “Burlo Garofolo”, 34137 Trieste, Italy; (R.P.); (M.C.)
| | | | - Elisabetta Fonzi
- Pharmacy Unit, S.Chiara Hospital, 38122 Trento, Italy; (E.F.); (A.P.)
| | | | - Stefano Vecchia
- Pharmacy Unit, Hospital Guglielmo da Saliceto, 29121 Piacenza, Italy;
| | - Marianna Veraldi
- Protesic and Pharmaceutical Assistance Sector n. 3, Department of Health Protection and Health Service Calabria Region, 88100 Catanzaro, Italy;
| | | | - Lucio Crinò
- Thoracic Oncology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (L.C.); (A.D.)
| | - Angelo Delmonte
- Thoracic Oncology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (L.C.); (A.D.)
| | - Carla Masini
- Pharmacy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| |
Collapse
|
18
|
Akers KG, Oskar S, Zhao B, Frederickson AM, Arunachalam A. Clinical Outcomes of PD-1/PD-L1 Inhibitors Among Patients With Advanced or Metastatic Non-Small Cell Lung Cancer With BRAF, ERBB2/HER2, MET , or RET Alterations: A Systematic Literature Review. J Immunother 2024; 47:128-138. [PMID: 38112201 PMCID: PMC10984634 DOI: 10.1097/cji.0000000000000500] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/21/2023] [Indexed: 12/21/2023]
Abstract
The therapeutic landscape for patients with advanced or metastatic non-small cell lung cancer (NSCLC) is rapidly evolving due to advances in molecular testing and the development of new targeted therapies and immunotherapies. However, the efficacy of programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) inhibitors in advanced or metastatic patients with NSCLC whose tumors harbor BRAF V600E mutation, HER2/ERBB2 alteration, MET exon 14 skipping mutation, or RET rearrangement is not completely understood. A systematic literature review was performed to summarize evidence from clinical trials and observational studies on objective response rate, progression-free survival, and overall survival in patients whose tumors express these biomarkers and who were treated with PD-1/PD-L1 inhibitors. Searches of Embase, MEDLINE, conference abstracts, and a clinical trial registry identified a total of 12 unique studies: 4 studies included patients with BRAF V600E mutation, 6 studies included patients with HER2/ERBB2 alteration, 7 studies included patients with MET exon 14 skipping mutation, and 5 studies included patients with RET rearrangement. Across studies, there was heterogeneity in treatment and patient characteristics and a lack of reporting on many important predictive and prognostic factors, including treatment regimens, patients' line of therapy, and tumor PD-L1 expression, which may explain the wide variation in objective response rate, progression-free survival, and overall survival across studies. Therefore, additional studies prospectively evaluating clinical outcomes of PD-1/PD-L1 inhibitors among patients with advanced or metastatic NSCLC whose tumors harbor emerging predictive or prognostic biomarkers are needed to determine whether this class of immunotherapy can provide additional survival benefits for these patients.
Collapse
Affiliation(s)
| | - Sabine Oskar
- Center for Observational and Real-World Evidence, Merck & Co., Inc., Rahway, NJ
| | - Bin Zhao
- Center for Observational and Real-World Evidence, Merck & Co., Inc., Rahway, NJ
| | | | - Ashwini Arunachalam
- Center for Observational and Real-World Evidence, Merck & Co., Inc., Rahway, NJ
| |
Collapse
|
19
|
Malapelle U, Passiglia F, Pepe F, Pisapia P, Lucia Reale M, Cortinovis D, Fraggetta F, Galetta D, Garbo E, Graziano P, Pagni F, Pasello G, Piovano P, Pilotto S, Tiseo M, Genova C, Righi L, Troncone G, Novello S. The biomarkers ATLAS: An audit on 1100 non-small cell lung cancer from an Italian knowledge-based database. Lung Cancer 2024; 191:107787. [PMID: 38593479 DOI: 10.1016/j.lungcan.2024.107787] [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: 12/27/2023] [Revised: 03/12/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024]
Abstract
AIMS To date, precision medicine has revolutionized the clinical management of Non-Small Cell Lung Cancer (NSCLC). International societies approved a rapidly improved mandatory testing biomarkers panel for the clinical stratification of NSCLC patients, but harmonized procedures are required to optimize the diagnostic workflow. In this context a knowledge-based database (Biomarkers ATLAS, https://biomarkersatlas.com/) was developed by a supervising group of expert pathologists and thoracic oncologists collecting updated clinical and molecular records from about 80 referral Italian institutions. Here, we audit molecular and clinical data from n = 1100 NSCLC patients collected from January 2019 to December 2020. METHODS Clinical and molecular records from NSCLC patients were retrospectively collected from the two coordinating institutions (University of Turin and University of Naples). Molecular biomarkers (KRAS, EGFR, BRAF, ROS1, ALK, RET, NTRK, MET) and clinical data (sex, age, histological type, smoker status, PD-L1 expression, therapy) were collected and harmonized. RESULTS Clinical and molecular data from 1100 (n = 552 mutated and n = 548 wild-type) NSCLC patients were systematized and annotated in the ATLAS knowledge-database. Molecular records from biomarkers testing were matched with main patients' clinical variables. CONCLUSIONS Biomarkers ATLAS (https://biomarkersatlas.com/) represents a unique, easily managing, and reliable diagnostic tool aiming to integrate clinical records with molecular alterations of NSCLC patients in the real-word Italian scenario.
Collapse
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Francesco Passiglia
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | | | - Diego Cortinovis
- Medical Oncology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | | | - Domenico Galetta
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Edoardo Garbo
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - Paolo Graziano
- Unit of Pathology, Scientific Institute for Research and Health Care (IRCCS) "Casa Sollievo della Sofferenza", San Giovanni Rotondo (FG), Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, University Milan Bicocca, Fondazione IRCCS San Gerardo, Monza, Italy
| | - Giulia Pasello
- Medical Oncology 2, Veneto Institute of Oncology (IOV) IRCCS, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Pierluigi Piovano
- SC Oncologia, Ospedale Ss. Antonio e Biagio e C. Arrigo, Alessandria, Italy
| | - Sara Pilotto
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust (AOUI) of Verona, Italy
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma and Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Carlo Genova
- UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova. Italy; Dipartimento di Medicina Interna e Specialità Mediche (DIMI), Università degli Studi di Genova, Italy
| | - Luisella Righi
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy
| | - Giancarlo Troncone
- Department of Public Health, Federico II University of Naples, Via S. Pansini, 5, 80131 Naples, Italy
| | - Silvia Novello
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Turin, Italy.
| |
Collapse
|
20
|
Scimone C, Pepe F, Russo G, Palumbo L, Ball G, Morel P, Russo A, Troncone G, Malapelle U. Technical evaluation of a novel digital PCR platform for detecting EGFR/KRAS mutations in NSCLC archived plasma specimens. THE JOURNAL OF LIQUID BIOPSY 2024; 3:100133. [PMID: 40026570 PMCID: PMC11863824 DOI: 10.1016/j.jlb.2023.100133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 03/05/2025]
Abstract
Introduction KRAS p.G12C hot spot mutations has rapidly modified diagnostic algorithm for lung cancer patients electing Non-Small Cell Lung Cancer (NSCLC) patients to target treatment. As regards, patients that harbor this hallmark showed a clinical benefit in terms of progression-free survival (PFS) and overall survival (OS) in comparison with control group under target treatment. In this scenario, KRAS p.G12C mutation requires optimized testing strategy in diagnostic routine practice. Although the widespread diffusion of NGS platforms, a not negligible percentage of Italian diagnostic institutions adopt singleplex technology (RT-PCR, dPCR) for molecular testing. Here, we aim to technically validate a novel dPCR system (QIAcuity™ Digital PCR System, Qiagen; Hilden, Germany) on a retrospective series of cfDNA samples from previously tested with a custom NGS system. (1,2)Methods: n = 50 liquid biopsy specimens (n = 25 KRAS/EGFR mutated and n = 25 wild type for actionable KRAS/EGFR mutations) from diagnostic routine NSCLC patients previously tested with a custom NGS panel were retrieved from our internal archival. Each sample was tested by adopting n = 5 KRAS and n = 3 EGFR commercially available dPCR assays on QIAcuity™ Digital PCR System (Qiagen; Hilden, Germany); an ultra-deep dPCR walk-away platform that automatizes molecular analysis. Technical sensitivity, technical specificity, and concordance rate between "gold standard" NGS system and QIAcuity™ Digital PCR System (Qiagen; Hilden, Germany) were assessed. Results Overall, all specimens were successfully analyzed with dPCR system. In details, 24 out of 25 mutated and 21 out of 24 wild type cases were detected. A technical sensitivity, specificity, and a concordance rate of 96.0 % (24/25), 88.0 % (22/25) and 92.0 % (46/50) were evaluated taking into account a MAF cut-off ≥ 0.2 % and a partition number of 100 positive partitions in wild-type channel. Conclusion Qiacuity (Qiagen; Hilden, Germany) platform enables accurate molecular analysis of diagnostic routine specimens. Optimized technical workflow is required to technically implement this platform in diagnostic routine setting.
Collapse
Affiliation(s)
- Claudia Scimone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Francesco Pepe
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Gianluca Russo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Lucia Palumbo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | | | | | | | - Giancarlo Troncone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| |
Collapse
|
21
|
Rekowska AK, Rola P, Kwiatkowska A, Wójcik-Superczyńska M, Gil M, Krawczyk P, Milanowski J. Abnormalities in the KRAS Gene and Treatment Options for NSCLC Patients with the G12C Mutation in This Gene-A Literature Review and Single-Center Experience. Biomedicines 2024; 12:325. [PMID: 38397927 PMCID: PMC10886466 DOI: 10.3390/biomedicines12020325] [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: 01/07/2024] [Revised: 01/22/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Mutations in the KRAS gene are among the most common mutations observed in cancer cells, but they have only recently become an achievable goal for targeted therapies. Two KRAS inhibitors, sotorasib and adagrasib, have recently been approved for the treatment of patients with advanced non-small cell lung cancer with the KRAS G12C mutation, while studies on their efficacy are still ongoing. In this work, we comprehensively analyzed RAS gene mutations' molecular background, mutation testing, KRAS inhibitors' effectiveness with an emphasis on non-small cell lung cancer, the impact of KRAS mutations on immunotherapy outcomes, and drug resistance problems. We also summarized ongoing trials and analyzed emerging perspectives on targeting KRAS in cancer patients.
Collapse
Affiliation(s)
- Anna K. Rekowska
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-090 Lublin, Poland (M.W.-S.); (M.G.); (J.M.)
| | | | | | | | | | - Paweł Krawczyk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-090 Lublin, Poland (M.W.-S.); (M.G.); (J.M.)
| | | |
Collapse
|
22
|
Sua LF, Osorio ÁE, Zuñiga-Restrepo V, Ibarra CD, Quintero N, Fernández-Trujillo L. Development of a Second Primary Lung Cancer Following a Primary Breast Cancer: A Case Series. J Investig Med High Impact Case Rep 2024; 12:23247096241272013. [PMID: 39390783 PMCID: PMC11468341 DOI: 10.1177/23247096241272013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 10/12/2024] Open
Abstract
Breast cancer (BC) accounts for 24.2% of all women's malignant tumors, with rising survival rates due to advancements in chemotherapy and targeted treatments. However, second primary cancers, particularly lung cancer (LC), have become more prevalent, often emerging approximately 10 years after BC treatment. This study presents a case series of 9 women diagnosed with second primary LC following BC, treated at a high-complexity hospital in Colombia between 2014 and 2019. All initial BCs were ductal carcinomas, 7 were triple negative, 1 was human epidermal growth factor receptor 2 positive, and 1 was estrogen and progesterone positive. Each patient had undergone radiation therapy, and 7 had received chemotherapy, increasing their LC risk. The second primary LCs, all adenocarcinomas, were confirmed using immunohistochemical stains for thyroid transcription factor-1 (TTF-1), Napsin A, and estrogen receptor (ER) status. The interval between treatments and LC detection ranged from 1 to 17 years, with 4 cases identified after 10 years and 3 within 1 to 3 years, underscoring the need for prolonged surveillance. Seven LCs were ipsilateral to the BC and radiation site, while 2 were contralateral, highlighting the necessity of monitoring both sides for potential LC development. This case series enhances the local epidemiological understanding, showing that prior radiotherapy for BC and histological analysis are key in characterizing second primary LC patients. The study emphasizes the critical role of accurate histological diagnosis in guiding treatment approaches for lung lesions in BC survivors.
Collapse
Affiliation(s)
- Luz F. Sua
- Fundación Valle del Lili, Cali, Colombia
- Universidad Icesi, Cali, Colombia
| | - Álvaro E. Osorio
- Fundación Valle del Lili, Cali, Colombia
- Universidad Icesi, Cali, Colombia
| | | | | | | | | |
Collapse
|
23
|
Harshbarger CL. Harnessing the power of Microscale AcoustoFluidics: A perspective based on BAW cancer diagnostics. BIOMICROFLUIDICS 2024; 18:011304. [PMID: 38434238 PMCID: PMC10907075 DOI: 10.1063/5.0180158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024]
Abstract
Cancer directly affects one in every three people, and mortality rates strongly correlate with the stage at which diagnosis occurs. Each of the multitude of methods used in cancer diagnostics has its own set of advantages and disadvantages. Two common drawbacks are a limited information value of image based diagnostic methods and high invasiveness when opting for methods that provide greater insight. Microfluidics offers a promising avenue for isolating circulating tumor cells from blood samples, offering high informational value at predetermined time intervals while being minimally invasive. Microscale AcoustoFluidics, an active method capable of manipulating objects within a fluid, has shown its potential use for the isolation and measurement of circulating tumor cells, but its full potential has yet to be harnessed. Extensive research has focused on isolating single cells, although the significance of clusters should not be overlooked and requires attention within the field. Moreover, there is room for improvement by designing smaller and automated devices to enhance user-friendliness and efficiency as illustrated by the use of bulk acoustic wave devices in cancer diagnostics. This next generation of setups and devices could minimize streaming forces and thereby enable the manipulation of smaller objects, thus aiding in the implementation of personalized oncology for the next generation of cancer treatments.
Collapse
Affiliation(s)
- C. L. Harshbarger
- Department of Orthopedics, Balgrist University Hospital, University of Zurich, Zurich, Switzerland; Institute for Biomechanics, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland; and Institute for Mechanical Systems, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| |
Collapse
|
24
|
Cerqueira ER, Batista PM, Almeida MF, Rego MAC, Ribeiro-Pereira ACP, Alencar F, Fernandes RA, Calabrich AFC, Schvartsman G. The journey of stage III and IV non-small cell lung cancer patients in the Brazilian private healthcare system: a retrospective study. Front Oncol 2023; 13:1257003. [PMID: 37920156 PMCID: PMC10619689 DOI: 10.3389/fonc.2023.1257003] [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: 07/17/2023] [Accepted: 09/07/2023] [Indexed: 11/04/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is still diagnosed at late stages in Brazil. The availability of newer treatment options has changed patient management, however, few real-world data have been published since then. This is a population-based retrospective cohort study that aims to evaluate the characteristics of stage III/IV NSCLC patients and their journey in the Brazilian private healthcare system. Patients aged ≥18 years, residing in Brazil who had their first medical appointment between 2016 and 2018 were included in the study. The sociodemographic and clinical characteristics of the patients and time intervals of interest were described. A total of 10,394 patients were analyzed. The majority of the patients were male (58.5%) with a median age of 64.0 (IQR = 58.0 - 71.0) years. In relation to characteristics of the disease, most of the tumors were characterized as adenocarcinomas (52.3%) and diagnosed at stage IV (72.2%). Most patients arrived at the hospital with an established NSCLC diagnosis, while 45.7% were diagnosed at the first medical appointment in the hospital or later. For patients who were diagnosed at the first medical appointment or later, a median interval of 15.0 (IQR = 6.0 - 33.0) days was observed between the first medical appointment and the diagnosis. The first treatment was given after a median of 25.0 (IQR = 6.0 - 49.0) days after diagnosis for patients without a prior diagnosis, and 57.0 (IQR: 33.0 - 98.0) days for patients with a prior diagnosis. The most common treatments were chemotherapy alone (33.8%), chemotherapy combined with radiotherapy (21.5%), radiotherapy alone (13.1%), adjuvant or neoadjuvant treatment (9.3%), surgery (3.3%), and immunotherapy (0.7%; alone or combined). At the end of follow-up (September, 2020), 52.3% of the patients had died. Despite having more treatment options in the private sector, data show that there is a need to improve access to technologies.
Collapse
Affiliation(s)
| | - Paula M. Batista
- Global Medical Scientific Affairs, MSD Brazil, São Paulo, Brazil
| | | | - Maria A. C. Rego
- Global Medical Scientific Affairs, MSD Brazil, São Paulo, Brazil
| | | | - Fernando Alencar
- Department of Health Economics, Origin Health, São Paulo, Brazil
| | | | | | - Gustavo Schvartsman
- Department of Medical Oncology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
25
|
Luo H, Wang W, Mai J, Yin R, Cai X, Li Q. The nexus of dynamic T cell states and immune checkpoint blockade therapy in the periphery and tumor microenvironment. Front Immunol 2023; 14:1267918. [PMID: 37881432 PMCID: PMC10597640 DOI: 10.3389/fimmu.2023.1267918] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/18/2023] [Indexed: 10/27/2023] Open
Abstract
Immune checkpoint blockade (ICB) therapies, that is, using monoclonal antibodies to reinvigorate tumor-reactive, antigen-specific T cells from the inhibitory effects of CTLA-4, PD-1 and PD-L1 immune checkpoints, have revolutionized the therapeutic landscape of modern oncology. However, only a subset of patients can benefit from the ICB therapy. Biomarkers associated with ICB response, resistance and prognosis have been subjected to intensive research in the past decade. Early studies focused on the analysis of tumor specimens and their residing microenvironment. However, biopsies can be challenging to obtain in clinical practice, and do not reflect the dynamic changes of immunological parameters during the ICB therapy. Recent studies have investigated profiles of antigen-specific T cells derived from the peripheral compartment using multi-omics approaches. By tracking the clonotype and diversity of tumor-reactive T cell receptor repertoire, these studies collectively establish that de novo priming of antigen-specific T cells in peripheral blood occurs throughout the course of ICB, whereas preexisting T cells prior to ICB are exhausted to various degrees. Here, we review what is known about ICB-induced T cell phenotypic and functional changes in cancer patients both within the tumor microenvironment and in the peripheral compartment. A better understanding of parameters influencing the response to ICBs will provide rationales for developing novel diagnostics and combinatorial therapeutic strategies to maximize the clinical efficacies of ICB therapies.
Collapse
Affiliation(s)
- Hong Luo
- Department of Obstetrics & Gynecology, Laboratory Medicine and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenxiang Wang
- Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, China
| | - Jia Mai
- Department of Obstetrics & Gynecology, Laboratory Medicine and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rutie Yin
- Department of Obstetrics & Gynecology, Laboratory Medicine and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xuyu Cai
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qintong Li
- Department of Obstetrics & Gynecology, Laboratory Medicine and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
26
|
Zia V, Lengyel CG, Tajima CC, de Mello RA. Advancements of ALK inhibition of non-small cell lung cancer: a literature review. Transl Lung Cancer Res 2023; 12:1563-1574. [PMID: 37577315 PMCID: PMC10413028 DOI: 10.21037/tlcr-22-619] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 06/19/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND AND OBJECTIVE The therapeutic landscape for non-small cell lung cancer (NSCLC) has evolved considerably in the last few years. The targeted drugs and molecular diagnostics have been developed together at a fast pace. This narrative review explores the evolution of anaplastic lymphoma kinase (ALK) targeting therapies from discovering the ALK protein, molecular tests, present clinical trial data and future perspectives. Since the body of evidence on lung cancer is growing daily, most oncologists need time to implement data in their daily practice. METHODS We developed a narrative review to provide up-to-date help in the clinical decision-making of ALK-altered NSCLC patients. In 2022, the authors reviewed PubMed's published pivotal randomized Phase 3 trial results. KEY CONTENT AND FINDINGS The development of ALK inhibitors was a revolution that is still ongoing; second and third-generation ALK inhibitors provided more than 30 months of progression-free survival (PFS) and impressive "brain-control". Brigatinib provided a survival benefit for patients with baseline brain metastases (HR 0.43, 95% CI: 0.21-0.89), and Lorlatinib demonstrated intracranial response rates of 82%, with 71% of complete intracranial responses. Personalized medicine is the new paradigm, from performing broad genetic panels for diagnosis to individual targeted therapy or combinations of different targeted agents. CONCLUSIONS In the future, performing broad molecular panels should be the standard of care in the front line and after each progression to detect arising resistance mechanisms. Longer PFS will substantially convert a deadly condition into an almost chronic disease in the following decades. Treatment sequencing will be the cornerstone for patient survival, and liquid biopsies may replace tissue biopsies.
Collapse
Affiliation(s)
- Victor Zia
- Post-graduation Program in Translational Medicine, University Federal of São Paulo, São Paulo, Brazil
| | - Csongor György Lengyel
- Department of Head and Neck Surgery, National Institute of Oncology Hungary, Budapest, Hungary
| | | | - Ramon Andrade de Mello
- Post-graduation Program in Translational Medicine, University Federal of São Paulo, São Paulo, Brazil
- Post-Graduation Programme in Medicine, Nine of July University (UNINOVE), São Paulo, Brazil
- Section of Medical Oncology, Brazilian Society of Cancerology (SBCANCER), Salvador, Brazil
- Department of Oncology, University of Oxford, Oxford, UK
- Oxford Cancer Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
27
|
Silas U, Blüher M, Bosworth Smith A, Saunders R. Fast In-House Next-Generation Sequencing in the Diagnosis of Metastatic Non-small Cell Lung Cancer: A Hospital Budget Impact Analysis. JOURNAL OF HEALTH ECONOMICS AND OUTCOMES RESEARCH 2023; 10:111-118. [PMID: 37389301 PMCID: PMC10306161 DOI: 10.36469/001c.77686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023]
Abstract
Background: Targeted therapy for cancer is becoming more frequent as the understanding of the molecular pathogenesis increases. Molecular testing must be done to use targeted therapy. Unfortunately, the testing turnaround time can delay the initiation of targeted therapy. Objective: To investigate the impact of a next-generation sequencing (NGS) machine in the hospital that would allow for in-house NGS testing of metastatic non-small cell lung cancer (mNSCLC) in a US setting. Methods: The differences between 2 hospital pathways were established with a cohort-level decision tree that feeds into a Markov model. A pathway that used in-house NGS (75%) and the use of external laboratories (so-called send-out NGS) (25%), was compared with the standard of exclusively send-out NGS. The model was from the perspective of a US hospital over a 5-year time horizon. All cost input data were in or inflated to 2021 USD. Scenario analysis was done on key variables. Results: In a hospital with 500 mNSCLC patients, the implementation of in-house NGS was estimated to increase the testing costs and the revenue of the hospital. The model predicted a $710 060 increase in testing costs, a $1 732 506 increase in revenue, and a $1 022 446 return on investment over 5 years. The payback period was 15 months with in-house NGS. The number of patients on targeted therapy increased by 3.38%, and the average turnaround time decreased by 10 days when in-house NGS was used. Discussion: Reducing testing turnaround time is a benefit of in-house NGS. It could contribute to fewer mNSCLC patients lost to second opinion and an increased number of patients on targeted therapy. The model outcomes predicted that, over a 5-year period, there would be a positive return on investment for a US hospital. The model reflects a proposed scenario. The heterogeneity of hospital inputs and the cost of send-out NGS means context-specific inputs are needed. Conclusion: Using in-house NGS testing could reduce the testing turnaround time and increase the number of patients on targeted therapy. Additional benefits for the hospital are that fewer patients will be lost to second opinion and that in-house NGS could generate additional revenue.
Collapse
Affiliation(s)
- Ubong Silas
- Coreva Scientific GmbH & Co. KG, Königswinter, Germany
| | | | | | | |
Collapse
|
28
|
Lazure P, Sireci A, Subbiah V, Murray S, Grohé C, Sherman SI, Kelly E, Bubach P, Péloquin S. Challenges in diagnosis and biomarker testing for RET-altered lung and thyroid cancer care: an international mixed-method study. BMC MEDICAL EDUCATION 2023; 23:410. [PMID: 37277734 DOI: 10.1186/s12909-023-04396-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 05/25/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND The introduction of new targeted therapies for RET-altered lung and thyroid cancers (LC/TC) has impacted pathologists' practice by making genomic testing more relevant. Variations in health systems and treatment access result in distinct clinical challenges and barriers. This study aimed to assess practice gaps and challenges experienced by pathologists involved in the diagnosis of RET-altered LC/TC, including biomarker testing, to inform educational solutions. METHODS Pathologists in Germany, Japan, the UK, and US participated in this ethics-approved mixed-methods study, which included interviews and surveys (data collected January-March 2020). Qualitative data was thematically analysed, quantitative data was analysed with chi-square and Kruskal-Wallis H-tests, and both were triangulated. RESULTS A total of 107 pathologists took part in this study. Knowledge gaps were reported regarding genomic testing for LC/TC in Japan (79/60%), the UK (73/66%), and the US (53/30%). Skill gaps were reported when selecting genomic biomarker tests to diagnose TC in Japan (79%), the UK (73%) and US (57%) and when performing specific biomarker tests, especially in Japan (82% for RET) and in the UK (75% for RET). Japanese participants (80%) reported uncertainty about what information to share with the multidisciplinary team to ensure optimal patient-centered care. At the time of data collection, pathologists in Japan faced access barriers to using RET biomarker tests: only 28% agreed that there are relevant RET genomic biomarker tests available in Japan, versus 67% to 90% in other countries. CONCLUSIONS This study identified areas where pathologists need additional continuing professional development opportunities to enhance their competencies and better support delivery of care to patients with RET-altered lung or thyroid tumours. Addressing identified gaps and improving competencies of pathologists in this field should be emphasised in continuing medical education curricula and through quality improvement initiatives. Strategies deployed on an institutional and health system level should aim to improve interprofessional communication and genetic biomarker testing expertise.
Collapse
Affiliation(s)
- Patrice Lazure
- AXDEV Group Inc., 8, Place du Commerce, Suite 210, Brossard, Québec, J4W 3H2, Canada.
| | | | - Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Suzanne Murray
- AXDEV Group Inc., 8, Place du Commerce, Suite 210, Brossard, Québec, J4W 3H2, Canada
| | | | - Steven I Sherman
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Sophie Péloquin
- AXDEV Group Inc., 8, Place du Commerce, Suite 210, Brossard, Québec, J4W 3H2, Canada
| |
Collapse
|
29
|
Osazuwa-Peters OL, Wilson LE, Check DK, Roberts MC, Srinivasan S, Clark AG, Crawford J, Chrischilles E, Carnahan RM, Campbell WS, Cowell LG, Greenlee R, Abbott AM, Mosa ASM, Mandhadi V, Stoddard A, Dinan MA. Factors Associated With Receipt of Molecular Testing and its Impact on Time to Initial Systemic Therapy in Metastatic Non-Small Cell Lung Cancer. Clin Lung Cancer 2023; 24:305-312. [PMID: 37055337 DOI: 10.1016/j.cllc.2023.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/13/2023] [Accepted: 03/10/2023] [Indexed: 04/15/2023]
Abstract
BACKGROUND Despite recommendations for molecular testing irrespective of patient characteristics, differences exist in receipt of molecular testing for oncogenic drivers amongst metastatic non-small cell lung cancer (mNSCLC) patients. Exploration into these differences and their effects on treatment is needed to identify opportunities for improvement. PATIENTS AND METHODS We conducted a retrospective cohort study of adult patients diagnosed with mNSCLC between 2011 and 2018 using PCORnet's Rapid Cycle Research Project dataset (n = 3600). Log-binomial, Cox proportional hazards (PH), and time-varying Cox regression models were used to ascertain whether molecular testing was received, and time from diagnosis to molecular testing and/or initial systemic treatment in the context of patient age, sex, race/ethnicity, and multiple comorbidities status. RESULTS The majority of patients in this cohort were ≤ 65 years of age (median [25th, 75th]: 64 [57, 71]), male (54.3%), non-Hispanic white individuals (81.6%), with > 2 comorbidities in addition to mNSCLC (54.1%). About half the cohort received molecular testing (49.9%). Patients who received molecular testing had a 59% higher probability of initial systemic treatment than patients who were yet to receive testing. Multiple comorbidity status was positively associated with receipt of molecular testing (RR, 1.27; 95% CI 1.08, 1.49). CONCLUSION Receipt of molecular testing in academic centers was associated with earlier initiation of systemic treatment. This finding underscores the need to increase molecular testing rates amongst mNSCLC patients during a clinically relevant period. Further studies to validate these findings in community centers are warranted.
Collapse
Affiliation(s)
| | - Lauren E Wilson
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC
| | - Devon K Check
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC
| | - Megan C Roberts
- Division of Pharmaceutical Outcomes and Policy, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC
| | - Swetha Srinivasan
- Division of Pharmaceutical Outcomes and Policy, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC
| | - Amy G Clark
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC
| | - Jeffrey Crawford
- Department of Medicine, Duke University School of Medicine, Durham, NC
| | | | - Ryan M Carnahan
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA
| | - W Scott Campbell
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - Lindsay G Cowell
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX
| | - Robert Greenlee
- Center for Clinical Epidemiology & Population Health, Marshfield Clinical Research Institute, Marshfield, WI
| | - Andrea M Abbott
- Department of Surgery, Medical University of South Carolina, Clinical Sciences, Charleston, SC
| | - Abu S M Mosa
- Department of Health Management and Informatics, University of Missouri School of Medicine, Columbia, MO
| | - Vasanthi Mandhadi
- Department of Health Management and Informatics, University of Missouri School of Medicine, Columbia, MO
| | - Alexander Stoddard
- Biomedical Informatics, Clinical and Translational Science Institute, Medical College of Wisconsin, Milwaukee, WI
| | - Michaela A Dinan
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC; Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT.
| |
Collapse
|
30
|
Burns L, Jani C, Radwan A, Omari OA, Patel M, Oxnard GR, Tapan U. Implementation Challenges and Disparities in Molecular Testing for Patients With Stage IV NSCLC: Perspectives from an Urban Safety-Net Hospital. Clin Lung Cancer 2023; 24:e69-e77. [PMID: 36464575 DOI: 10.1016/j.cllc.2022.11.002] [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: 06/29/2022] [Revised: 10/27/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022]
Abstract
The advent of next-generation sequencing (NGS), including both tissue assays and circulating tumor DNA (ct-DNA), has been pivotal in improving outcomes for patients with non-small cell lung cancer (NSCLC). Although molecular testing is standard of care for advanced NSCLC, challenges still exist in its implementation. This Perspective examines barriers to the widespread implementation of NGS from the vantage point of a single urban safety-net institution, with a particular focus on examining racial disparities in NGS completion. We conducted a review of patients at our institution from January 2015 through January 2022 and examined molecular testing patterns before and after the publication of updated molecular testing guidelines from the International Association for the Study of Lung Cancer (IASLC), Association for Molecular Pathology (AMP), and College of American Pathologists (CAP) in March of 2018. While NGS increased over time, we found that 43% of patients in the March 2018 through January 2022 group still did not receive NGS, and the most common reasons for the absence of testing included a lack of physician ordering and insufficient tissue on biopsy. We did not note any racial disparities in completion or time-to-adoption of NGS. Patients with squamous cell carcinoma (SCC) histology were noted to receive liquid NGS markedly less often than patients with non-squamous histology in the March 2018 through January 2022 period. Based on our own data and a review of findings from colleagues in the field, we advocate for additional physician educational programming, increased use of ct-DNA biopsy, automated (reflexive) NGS tissue testing on receipt of biopsy, and consideration for the broader molecular profiling of patients with SCC histology.
Collapse
Affiliation(s)
- Laura Burns
- Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA.
| | - Chinmay Jani
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA
| | - Amr Radwan
- Section of Hematology & Medical Oncology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA
| | - Omar Al Omari
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA
| | - Mohini Patel
- Boston University School of Public Health and Boston Medical Center, Boston, MA
| | - Geoffrey R Oxnard
- Section of Hematology & Medical Oncology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA
| | - Umit Tapan
- Section of Hematology & Medical Oncology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA
| |
Collapse
|
31
|
Bessi S, Pepe F, Russo G, Pisapia P, Ottaviantonio M, Biancalani F, Iaccarino A, Russo M, Biancalani M, Troncone G, Malapelle U. Comparison of two next-generation sequencing-based approaches for liquid biopsy analysis in patients with non-small cell lung cancer: a multicentre study. J Clin Pathol 2023; 76:206-210. [PMID: 35701144 DOI: 10.1136/jclinpath-2022-208308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/31/2022] [Indexed: 11/03/2022]
Abstract
In the era of personalised medicine, testing for an increasing number of predictive biomarkers is becoming a priority. However, tissue biopsies from these patients are oftentimes insufficient for conventional approaches, a common issue that deprives them of the clinical benefits of biomarker-directed treatments. To tackle this problem, many clinical laboratories are resorting to circulating tumour DNA (ctDNA), which is becoming increasingly appreciated as a valuable source for biomarker testing. In this context, next-generation sequencing (NGS) has become essential. Indeed, different NGS systems are able to detect several clinically relevant low-frequency hot-spot mutations simultaneously in a single run. However, their reproducibility in the analysis of ctDNA has not yet been investigated. The purpose of this study was to evaluate the reproducibility of using Illumina MiSeq and Thermo Fisher Ion S5 Plus platforms to assess pathogenic alterations in non-small cell lung cancer (NSCLC) liquid biopsy specimens. Using the in vitro diagnostic (IVD) NGS panel Myriapod NGS Cancer panel DNA (Diatech Pharmacogenetics) on MiSeq platform (Illumina), we reanalysed ctDNA extracted from a retrospective series of n=40 patients with advanced NSCLC previously tested with a custom NGS panel (SiRe) on Thermo Fisher Ion S5 Plus system. Overall, 13 out of 40 (32.5%) ctDNA samples displayed pathogenic alterations in at least two genes, namely, EGFR and KRAS A concordance rate of 100% was identified between the two methodologies in terms of sample mutational status and total number of detected variables. All NGS platforms featured a high degree of concordance.
Collapse
Affiliation(s)
- Silvia Bessi
- Departmental Structure of Oncological Molecular Pathology, Azienda USL Toscana Centro, Prato, Italy
| | - Francesco Pepe
- Public Health, University of Naples Federico II, Naples, Italy
| | - Gianluca Russo
- Public Health, University of Naples Federico II, Naples, Italy
| | | | - Marco Ottaviantonio
- Departmental Structure of Oncological Molecular Pathology, Azienda USL Toscana Centro, Prato, Italy
| | | | | | - Maria Russo
- Public Health, University of Naples Federico II, Naples, Italy
| | - Mauro Biancalani
- Morphological Diagnostic and Biomolecular Characterization Area, Complex Unit of Pathological Anatomy, Azienda USL Toscana Centro, Prato, Italy
| | | | | |
Collapse
|
32
|
Behnke A, Cayre A, De Maglio G, Giannini G, Habran L, Tarsitano M, Chetta M, Cappellen D, Lespagnol A, Le Naoures C, Massazza G, Destro A, Bonzheim I, Rau A, Battmann A, Kah B, Watkin E, Hummel M. FACILITATE: A real-world, multicenter, prospective study investigating the utility of a rapid, fully automated real-time PCR assay versus local reference methods for detecting epidermal growth factor receptor variants in NSCLC. Pathol Oncol Res 2023; 29:1610707. [PMID: 36798672 PMCID: PMC9927408 DOI: 10.3389/pore.2023.1610707] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023]
Abstract
Accurate testing for epidermal growth factor receptor (EGFR) variants is essential for informing treatment decisions in non-small cell lung cancer (NSCLC). Automated diagnostic workflows may allow more streamlined initiation of targeted treatments, where appropriate, while comprehensive variant analysis is ongoing. FACILITATE, a real-world, prospective, multicenter, European study, evaluated performance and analytical turnaround time of the Idylla™ EGFR Mutation Test compared with local reference methods. Sixteen sites obtained formalin-fixed paraffin-embedded biopsy samples with ≥ 10% neoplastic cells from patients with NSCLC. Consecutive 5 μm sections from patient samples were tested for clinically relevant NSCLC-associated EGFR variants using the Idylla™ EGFR Mutation Test and local reference methods; performance (concordance) and analytical turnaround time were compared. Between January 2019 and November 2020, 1,474 parallel analyses were conducted. Overall percentage agreement was 97.7% [n = 1,418; 95% confidence interval (CI): 96.8-98.3], positive agreement, 87.4% (n = 182; 95% CI: 81.8-91.4) and negative agreement, 99.2% (n = 1,236; 95% CI: 98.5-99.6). There were 38 (2.6%) discordant cases. Ninety percent of results were returned with an analytical turnaround time of within 1 week using the Idylla™ EGFR Mutation Test versus ∼22 days using reference methods. The Idylla™ EGFR Mutation Test performed well versus local methods and had shorter analytical turnaround time. The Idylla™ EGFR Mutation Test can thus support application of personalized medicine in NSCLC.
Collapse
Affiliation(s)
- Anke Behnke
- Charité-Universitätsmedizin Berlin, Institute of Pathology and Berlin Institute of Health, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anne Cayre
- Département de Pathologie, Centre Jean-Perrin, Clermont-Ferrand, France
| | - Giovanna De Maglio
- Azienda Sanitaria Universitaria Friuli Centrale, Pathology Department, Santa Maria della Misericordia Hospital, Udine, Italy
| | - Giuseppe Giannini
- Department Molecular Medicine, Università di Roma La Sapienza, Rome, Italy
| | - Lionel Habran
- Anatomopathology Department, CHU Liège, Liège, Belgium
| | - Marina Tarsitano
- Di Laboratorio, A.O.R.N. Cardarelli, Medical Genetics Laboratory, and Ospedale Antonio Cardarelli, U.O.C. di Genetica Medica, Naples, Italy
| | - Massimiliano Chetta
- Di Laboratorio, A.O.R.N. Cardarelli, Medical Genetics Laboratory, and Ospedale Antonio Cardarelli, U.O.C. di Genetica Medica, Naples, Italy
| | - David Cappellen
- Service de Biologie des Tumeurs, Centre Hospitalier Universitaire de Bordeaux, Hôpital du Haut Lévêque, Pessac, France
| | - Alexandra Lespagnol
- CHU de Rennes, Laboratoire de Génétique Somatique des Cancers, Rennes, France
| | - Cecile Le Naoures
- CHU de Rennes, Service d’Anatomie et Cytologie Pathologiques, Rennes, France
| | - Gabriella Massazza
- Dipartimento Medicina di Laboratorio Anatomia Patologica, ASST Papa Giovanni XXIII, Bergamo, BG, Italy
| | - Annarita Destro
- Pathology Department, Humanitas Clinical and Research Center—IRCCS, Milan, Italy
| | - Irina Bonzheim
- Institute of Pathology and Neuropathology, Eberhard Karls University of Tübingen and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - Achim Rau
- Institute of Pathology and Neuropathology, Eberhard Karls University of Tübingen and Comprehensive Cancer Center, University Hospital Tübingen, Tübingen, Germany
| | - Achim Battmann
- Institut für Pathologie und Zytodiagnostik am Krankenhaus Nordwest, Frankfurt, Germany
| | - Bettina Kah
- Institut für Hämatopathologie Hamburg, Hamburg, Germany
| | | | - Michael Hummel
- Charité-Universitätsmedizin Berlin, Institute of Pathology and Berlin Institute of Health, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany,*Correspondence: Michael Hummel,
| |
Collapse
|
33
|
Fintelmann FJ, Martin NA, Tahir I, Quinn EM, Allen TC, Joseph L, Nikolic B, Lee C. Optimizing molecular testing of lung cancer needle biopsy specimens: potential solutions from an interdisciplinary qualitative study. Respir Res 2023; 24:17. [PMID: 36650544 PMCID: PMC9847026 DOI: 10.1186/s12931-023-02321-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Molecular testing can detect actionable genomic alterations and tumor cell surface proteins in patients with non-small cell lung cancer (NSCLC). However, utilization remains suboptimal, representing missed treatment opportunities. This study aimed to identify challenges and potential solutions to obtaining percutaneous lung needle biopsy specimens for successful molecular testing in patients with advanced NSCLC. METHODS This interdisciplinary qualitative study included ten radiologists and four pathologists from academic and community settings across the United States who routinely perform and analyze percutaneous lung needle biopsies. Participants underwent semi-structured one-on-one interviews (Phase 1). Interview questionnaires were constructed based on a literature review of key lines of inquiry and conducted by professional market researchers using the theoretical domains framework. Primary barriers to molecular testing were identified using thematic analysis. Subsequently, multidisciplinary focus groups were convened to identify potential solutions (Phase 2). RESULTS Four themes emerged as barriers to molecular testing and were matched to the clinical workflow: (1) biopsy request, (2) biopsy procedure, (3) specimen analysis, and (4) communication. The nineteen potential solutions included adding a "checkbox" to indicate molecular testing in the biopsy request, leveraging pre-procedural imaging to guide biopsies, conserving tissue through appropriate allocation strategies and next generation sequencing panels instead of sequential single-gene assays, instituting reflex-molecular testing upon NSCLC diagnosis, tracking and communicating biopsy outcomes at multidisciplinary tumor boards, and improving integration of radiologists and pathologists into oncology care teams. CONCLUSIONS Potential solutions exist to increase successful molecular testing of lung needle biopsy specimens in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Florian J. Fintelmann
- grid.32224.350000 0004 0386 9924Department of Radiology, Division of Thoracic Imaging and Intervention, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| | - Nikki A. Martin
- grid.443873.f0000 0004 0422 4933LUNGevity Foundation, Bethesda, MD USA
| | - Ismail Tahir
- grid.32224.350000 0004 0386 9924Department of Radiology, Division of Thoracic Imaging and Intervention, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| | - Elissa M. Quinn
- grid.497611.c0000 0004 1794 1958Blueprint Medicines, Boston, MA USA
| | | | - Lija Joseph
- grid.461527.30000 0004 0383 4123Lowell General Hospital, Lowell, MA USA
| | - Boris Nikolic
- grid.439147.c0000 0004 0628 7583Wyoming Valley Radiology Associates, Wilkes-Barre General Hospital, Wilkes-Barre, PA USA
| | - Christopher Lee
- grid.50956.3f0000 0001 2152 9905Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, CA USA
| |
Collapse
|
34
|
Thurfjell V, Micke P, Yu H, Krupar R, Svensson MA, Brunnström H, Lamberg K, Moens LNJ, Strell C, Gulyas M, Helenius G, Yoshida A, Goldmann T, Mattsson JSM. Comparison of ROS1-rearrangement detection methods in a cohort of surgically resected non-small cell lung carcinomas. Transl Lung Cancer Res 2022; 11:2477-2494. [PMID: 36636421 PMCID: PMC9830269 DOI: 10.21037/tlcr-22-504] [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: 07/04/2022] [Accepted: 11/06/2022] [Indexed: 12/14/2022]
Abstract
Background Patients with non-small cell lung cancer (NSCLC) harboring a ROS proto-oncogene 1 (ROS1)-rearrangement respond to treatment with ROS1 inhibitors. To distinguish these rare cases, screening with immunohistochemistry (IHC) for ROS1 protein expression has been suggested. However, the reliability of such an assay and the comparability of the antibody clones has been debated. Therefore we evaluated the diagnostic performance of current detection strategies for ROS1-rearrangement in two NSCLC-patient cohorts. Methods Resected tissue samples, retrospectively collected from consecutive NSCLC-patients surgically treated at Uppsala University Hospital were incorporated into tissue microarrays [all n=676, adenocarcinomas (AC) n=401, squamous cell carcinomas (SCC) n=213, other NSCLC n=62]. ROS1-rearrangements were detected using fluorescence in situ hybridization (FISH) (Abbott Molecular; ZytoVision). In parallel, ROS1 protein expression was detected using IHC with three antibody clones (D4D6, SP384, EPMGHR2) and accuracy, sensitivity, and specificity were determined. Gene expression microarray data (Affymetrix) and RNA-sequencing data were available for a subset of patients. NanoString analyses were performed for samples with positive or ambiguous results (n=21). Results Using FISH, 2/630 (0.3% all NSCLC; 0.5% non-squamous NSCLC) cases were positive for ROS1 fusion. Additionally, nine cases demonstrated ambiguous FISH results. Using IHC, ROS1 protein expression was detected in 24/665 (3.6% all NSCLC; 5.1% non-squamous NSCLC) cases with clone D4D6, in 18/639 (2.8% all NSCLC; 3.9% non-squamous NSCLC) cases with clone SP384, and in 1/593 (0.2% all NSCLC; 0.3% non-squamous NSCLC) case with clone EPMGHR2. Elevated RNA-levels were seen in 19/369 (5.1%) cases (Affymetrix and RNA-sequencing combined). The overlap of positive results between the assays was poor. Only one of the FISH-positive cases was positive with all antibodies and demonstrated high RNA-expression. This rearrangement was confirmed in the NanoString-assay and also in the RNA-sequencing data. Other cases with high protein/RNA-expression or ambiguous FISH were negative in the NanoString-assay. Conclusions The occurrence of ROS1 fusions is low in our cohorts. The IHC assays detected the fusions, but the accuracy varied depending on the clone. The presumably false-positive and uncertain FISH results questions this method for detection of ROS1-rearrangements. Thus, when IHC is used for screening, transcript-based assays are preferable for validation in clinical diagnostics.
Collapse
Affiliation(s)
- Viktoria Thurfjell
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hui Yu
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Rosemarie Krupar
- Division of Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany;,Institute of Pathology, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Maria A. Svensson
- Clinical Research Center, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Hans Brunnström
- Division of Pathology, Lund University and Laboratory Medicine Region Skåne, Lund, Sweden
| | - Kristina Lamberg
- Department of Pulmonary and Allergic Diseases, Uppsala University Hospital, Uppsala, Sweden
| | - Lotte N. J. Moens
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden;,Clinical Genomics Uppsala, Science for Life Laboratory, Uppsala, Sweden
| | - Carina Strell
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Miklos Gulyas
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Gisela Helenius
- Department of Laboratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Akihiko Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Torsten Goldmann
- Division of Pathology, Research Center Borstel, Leibniz Lung Center, Borstel, Germany;,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Großhansdorf, Germany
| | | |
Collapse
|
35
|
Liquid Biopsy in the Oncological Management of a Histologically Undiagnosed Lung Carcinoma: A Case Report. J Pers Med 2022; 12:jpm12111874. [PMID: 36579578 PMCID: PMC9694216 DOI: 10.3390/jpm12111874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022] Open
Abstract
Lung cancer is one of the most common and lethal cancers worldwide. Numerous medications targeting specific molecular alterations in non-small cell lung cancer have been introduced in the last decade and have revolutionized the clinical management of the disease. Their use has brought to a parallel evolution of molecular testing techniques to identify alterations in druggable molecular targets within the genetic material of the tumors. To perform molecular testing, biopsy or surgery tissue specimens are needed, which in addition allow the histological characterization of the tumors. Unfortunately, in real-life practice not all the patients are suitable for biopsy or surgery procedures. The use of liquid biopsy for blood extracted tumoral DNA analysis is a promising approach in unbiopsied cases, but it is also weighted by several methodological and technical limitations. We report here a case of histologically undiagnosed lung cancer managed with a liquid biopsy and subsequently with anti-EGFR treatment. Our report highlights that the use of liquid biopsy molecular testing in specific clinical situations can offer treatment opportunities for fragile patients affected by lung cancer.
Collapse
|
36
|
Lai GGY, Guo R, Drilon A, Shao Weng Tan D. Refining patient selection of MET-activated non-small cell lung cancer through biomarker precision. Cancer Treat Rev 2022; 110:102444. [PMID: 36108503 PMCID: PMC10961969 DOI: 10.1016/j.ctrv.2022.102444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 07/21/2022] [Accepted: 07/28/2022] [Indexed: 12/12/2022]
Abstract
Dysregulated MET signaling plays an important role in lung oncogenesis, tumor growth and invasiveness. It may occur through various mechanisms, such as MET overexpression or gene amplification or mutation, all of which can be detected by specific methods. The utility of MET overexpression as a biomarker remains unclear due to discrepancies in its occurrence and non-standardized cut-off thresholds. MET exon 14 skipping mutation (METex14) was established as a strong predictor of response to selective MET tyrosine kinase inhibitors (TKIs), and clinical trial results in patients with non-small cell lung cancer (NSCLC) harboring METex14 led to the approval of capmatinib and tepotinib by regulatory agencies worldwide. MET amplification is an emerging biomarker, with clinical data indicating an association between MET gene copy number and response to MET-TKIs. Additionally, MET amplification represents an important mechanism of resistance to TKIs in oncogene-driven NSCLC. The identification of molecular alterations for which targeted therapies are available is important, and high-throughput next-generation sequencing techniques can provide information on multiple genes at the same time, helping to provide valuable predictive information for oncogene-driven cancers. This review summarizes the current methods used for the detection of METex14, MET amplification and MET overexpression, and discusses the evidence for the use of MET-TKIs in patients with NSCLC with MET dysregulation. We discuss the practical challenges that impact the use of METex14 in the clinic and the evidence gaps that need to be addressed to validate additional genomic markers for MET-dependent cancers.
Collapse
Affiliation(s)
- Gillianne G Y Lai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Robin Guo
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | | |
Collapse
|
37
|
Pujol N, Heeke S, Bontoux C, Boutros J, Ilié M, Hofman V, Marquette CH, Hofman P, Benzaquen J. Molecular Profiling in Non-Squamous Non-Small Cell Lung Carcinoma: Towards a Switch to Next-Generation Sequencing Reflex Testing. J Pers Med 2022; 12:1684. [PMID: 36294823 PMCID: PMC9605324 DOI: 10.3390/jpm12101684] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/07/2022] [Accepted: 10/08/2022] [Indexed: 11/05/2022] Open
Abstract
Molecular diagnosis of lung cancer is a constantly evolving field thanks to major advances in precision oncology. The wide range of actionable molecular alterations in non-squamous non-small cell lung carcinoma (NS-NSCLC) and the multiplicity of mechanisms of resistance to treatment resulted in the need for repeated testing to establish an accurate molecular diagnosis, as well as to track disease evolution over time. While assessing the increasing complexity of the molecular composition of tumors at baseline, as well as over time, has become increasingly challenging, the emergence and implementation of next-generation sequencing (NGS) testing has extensively facilitated molecular profiling in NS-NSCLC. In this review, we discuss recent developments in the molecular profiling of NS-NSCLC and how NGS addresses current needs, as well as how it can be implemented to address future challenges in the management of NS-NSCLC.
Collapse
Affiliation(s)
- Nina Pujol
- Centre Antoine-Lacassagne, Department of Radiation Oncology, Côte d’Azur University, 06000 Nice, France
| | - Simon Heeke
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christophe Bontoux
- Laboratory of Clinical and Experimental Pathology, Côte d’Azur University, Pasteur 1 Hospital, Centre Hospitalier Universitaire de Nice, FHU OncoAge, Biobank BB-0033-00025, 06000 Nice, France
- CNRS UMR 7284, INSERM U1081, Institute of Research on Cancer and Aging, Côte d’Azur University, 06000 Nice, France
| | - Jacques Boutros
- CNRS UMR 7284, INSERM U1081, Institute of Research on Cancer and Aging, Côte d’Azur University, 06000 Nice, France
- Department of Pulmonary Medicine and Thoracic Oncology, Côte d’Azur University, Pasteur 1 Hospital, Centre Hospitalier Universitaire de Nice, FHU OncoAge, 06000 Nice, France
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Côte d’Azur University, Pasteur 1 Hospital, Centre Hospitalier Universitaire de Nice, FHU OncoAge, Biobank BB-0033-00025, 06000 Nice, France
- CNRS UMR 7284, INSERM U1081, Institute of Research on Cancer and Aging, Côte d’Azur University, 06000 Nice, France
| | - Véronique Hofman
- Laboratory of Clinical and Experimental Pathology, Côte d’Azur University, Pasteur 1 Hospital, Centre Hospitalier Universitaire de Nice, FHU OncoAge, Biobank BB-0033-00025, 06000 Nice, France
- CNRS UMR 7284, INSERM U1081, Institute of Research on Cancer and Aging, Côte d’Azur University, 06000 Nice, France
| | - Charles-Hugo Marquette
- CNRS UMR 7284, INSERM U1081, Institute of Research on Cancer and Aging, Côte d’Azur University, 06000 Nice, France
- Department of Pulmonary Medicine and Thoracic Oncology, Côte d’Azur University, Pasteur 1 Hospital, Centre Hospitalier Universitaire de Nice, FHU OncoAge, 06000 Nice, France
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Côte d’Azur University, Pasteur 1 Hospital, Centre Hospitalier Universitaire de Nice, FHU OncoAge, Biobank BB-0033-00025, 06000 Nice, France
- CNRS UMR 7284, INSERM U1081, Institute of Research on Cancer and Aging, Côte d’Azur University, 06000 Nice, France
| | - Jonathan Benzaquen
- CNRS UMR 7284, INSERM U1081, Institute of Research on Cancer and Aging, Côte d’Azur University, 06000 Nice, France
- Department of Pulmonary Medicine and Thoracic Oncology, Côte d’Azur University, Pasteur 1 Hospital, Centre Hospitalier Universitaire de Nice, FHU OncoAge, 06000 Nice, France
| |
Collapse
|
38
|
Navani N, Butler R, Ibrahimo S, Verma A, Evans M, Doherty GJ, Ahmed S. Optimising tissue acquisition and the molecular testing pathway for patients with non-small cell lung cancer: A UK expert consensus statement. Lung Cancer 2022; 172:142-153. [PMID: 36099709 DOI: 10.1016/j.lungcan.2022.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 11/21/2022]
Abstract
Targeted therapy against actionable variants has revolutionised the treatment landscape for non-small cell lung cancer (NSCLC). Approximately half of NSCLC adenocarcinomas have an actionable variant, making molecular testing a critical component of the diagnostic process to personalise therapeutic options, optimise clinical outcomes and minimise toxicity. Recently, genomic testing in England has undergone major changes with the introduction of Genomic Laboratory Hubs, designed to consolidate and enhance existing laboratory provision and deliver genomic testing as outlined in the National Genomic Test Directory. Similar changes are ongoing in Scotland, Wales and Northern Ireland. However, multiple challenges exist with current tissue acquisition procedures and the molecular testing pathway in the UK, including quantity and quality of available tissue, adequacy rates, test availability among genomic laboratories, turnaround times, multidisciplinary team communication, and limited guidance and standardisation. The COVID-19 pandemic has added an extra layer of complexity. Herein, we summarise best practice recommendations, based on expert opinion, to overcome existing challenges in the UK. The least invasive biopsy technique should be undertaken with the aim of acquiring the greatest quality and quantity of tissue. Use of sedation should be considered to improve patient experience. Rapid on-site evaluation may also be useful to help guide adequate sampling, and liquid biopsy may be beneficial in some instances. Sample processing should be appropriate to facilitate biomarker testing, in particular, next-generation sequencing for comprehensive genomic information. Steps to optimise tissue utilisation and turnaround times, such as planning of tissue usage, limiting immunohistochemistry, tumour enrichment, and reflex testing at diagnosis, should be implemented. Guidelines for tissue acquisition and sample processing may help to improve sample adequacy to perform downstream testing. Communication among genomic laboratories will help to standardise test availability across England and local auditing could identify further areas for optimisation, including ways to improve turnaround times and adequacy rates.
Collapse
Affiliation(s)
- Neal Navani
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, United Kingdom; University College London Hospitals NHS Foundation Trust, London, United Kingdom.
| | - Rachel Butler
- North Thames Genomic Laboratory Hub, Great Ormond Street Hospital, London, United Kingdom
| | | | | | - Matthew Evans
- Black Country Pathology Services, West Midlands, United Kingdom
| | - Gary J Doherty
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Samreen Ahmed
- University Hospitals of Leicester NHS Trust, Leicester, United Kingdom
| |
Collapse
|
39
|
Pisapia P, L'Imperio V, Galuppini F, Sajjadi E, Russo A, Cerbelli B, Fraggetta F, d'Amati G, Troncone G, Fassan M, Fusco N, Pagni F, Malapelle U. The evolving landscape of anatomic pathology. Crit Rev Oncol Hematol 2022; 178:103776. [PMID: 35934262 DOI: 10.1016/j.critrevonc.2022.103776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/11/2022] Open
Abstract
Anatomic pathology has changed dramatically in recent years. Although the microscopic assessment of tissues and cells is and will remain the mainstay of cancer diagnosis molecular profiling has become equally relevant. Thus, to stay abreast of the evolving landscape of today's anatomic pathology, modern pathologists must be able to master the intricate world of predictive molecular pathology. To this aim, pathologists have had to acquire additional knowledge to bridge the gap between clinicians and molecular biologists. This new role is particularly important, as cases are now collegially discussed in molecular tumor boards (MTBs). Moreover, as opposed to traditional pathologists, modern pathologists have also adamantly embraced innovation while keeping a constant eye on tradition. In this article, we depict the highlights and shadows of the upcoming "Anatomic Pathology 2.0" by placing particular emphasis on the pathologist's growing role in the management of cancer patients.
Collapse
Affiliation(s)
- Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Vincenzo L'Imperio
- Department of Medicine and Surgery, Pathology, University of Milan-Bicocca (UNIMIB), Monza, Italy
| | - Francesca Galuppini
- Unit of Surgical Pathology, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | - Bruna Cerbelli
- Department of Radiology, Oncology and Pathology, Sapienza, University of Rome, Rome, Italy
| | - Filippo Fraggetta
- Pathology Unit, Gravina Hospital Caltagirone, ASP Catania, Caltagirone, Italy
| | - Giulia d'Amati
- Department of Radiology, Oncology and Pathology, Sapienza, University of Rome, Rome, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Matteo Fassan
- Unit of Surgical Pathology, Department of Medicine (DIMED), University of Padua, Padua, Italy; Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Veneto, Italy.
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, University of Milan-Bicocca (UNIMIB), Monza, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
40
|
Danesi V, Massa I, Foca F, Delmonte A, Crinò L, Bronte G, Ragonesi M, Maltoni R, Manunta S, Cravero P, Andrikou K, Priano I, Balzi W, Gentili N, Burke T, Altini M. Real-World Outcomes and Treatments Patterns Prior and after the Introduction of First-Line Immunotherapy for the Treatment of Metastatic Non-Small Cell Lung Cancer. Cancers (Basel) 2022; 14:cancers14184481. [PMID: 36139641 PMCID: PMC9497168 DOI: 10.3390/cancers14184481] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/26/2022] Open
Abstract
Simple Summary The advent of immuno-oncology (IO) agents, particularly immune checkpoint inhibitors (ICIs), has changed the treatment landscape of non-small cell lung cancer (NSCLC). We performed a retro-prospective study to describe the patients’ outcomes prior to and after the local regulatory approval of pembrolizumab as a first-line (1L) treatment in the real-world setting of an Italian cancer centre. Analyses were performed of a total of 694 patients with no or unknown oncogene addicted tumour, grouped into Pre- (n = 344) and Post- (n = 350) 1L IO populations. The study provides evidence of improvements in overall survival associated with the introduction of 1L immunotherapy, suggesting that receiving immunotherapy in the first-line rather than in the second- or later lines of treatment may be more favourable. Abstract Background: This study provides insights into the treatment use and outcomes of metastatic non-small cell lung cancer (NSCLC) patients in a real-world setting prior to and after the availability of immuno-oncology (IO) regimens in the first line (1L). Methods: Metastatic NSCLC patients, who initiated systemic 1L anticancer treatment from 2014 to 2020, were identified from health records. Patients were grouped into Pre-1L IO and Post-1L IO, according to the availability of pembrolizumab 1L monotherapy at the date of initiating 1L systemic anticancer treatment. Patient characteristics, treatment patterns and outcomes were assessed by the cohort. Overall survival (OS) and real-world progression-free survival (rwPFS) were calculated using the Kaplan-Meier method. Results: The most common 1L treatment was platinum-based chemotherapy regimens in both groups (≥46%), followed by single-agent chemotherapy (27.0%) in Pre-1L IO and pembrolizumab (26.0%) in Post-1L IO. Median OS was 6.2 (95% CI 5.5–7.4) in Pre- and 8.9 months (95% CI 7.5–10.6) in Post-1L IO, while rwPFS was 3.7 (95% CI 3.3–4.2) and 4.7 months (95% CI 3.9–5.7), respectively. Conclusions: Even if a small proportion of patients received a 1L IO, the data showed an improved survival outcomes in the Post-1L IO group.
Collapse
Affiliation(s)
- Valentina Danesi
- Outcome Research, Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Ilaria Massa
- Outcome Research, Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Flavia Foca
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
- Correspondence:
| | - Angelo Delmonte
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Lucio Crinò
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Giuseppe Bronte
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Maria Ragonesi
- Nursing Service, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Roberta Maltoni
- Outcome Research, Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Silvia Manunta
- AULSS5, UOC Oncologia, Ospedale Santa Maria della Misericordia, 45100 Rovigo, Italy
| | - Paola Cravero
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Kalliopi Andrikou
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Ilaria Priano
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - William Balzi
- Outcome Research, Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Nicola Gentili
- Outcome Research, Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy
| | - Thomas Burke
- Center for Observational and Real World Evidence, Merck & Co Inc., Kenilworth, NJ 07033, USA
| | - Mattia Altini
- Healthcare Administration, Azienda Unità Sanitaria Locale della Romagna, 48121 Ravenna, Italy
| |
Collapse
|
41
|
Horgan D, Čufer T, Gatto F, Lugowska I, Verbanac D, Carvalho Â, Lal JA, Kozaric M, Toomey S, Ivanov HY, Longshore J, Malapelle U, Hasenleithner S, Hofman P, Alix-Panabières C. Accelerating the Development and Validation of Liquid Biopsy for Early Cancer Screening and Treatment Tailoring. Healthcare (Basel) 2022; 10:1714. [PMID: 36141326 PMCID: PMC9498805 DOI: 10.3390/healthcare10091714] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 11/26/2022] Open
Abstract
Liquid biopsy (LB) is a minimally invasive method which aims to detect circulating tumor-derived components in body fluids. It provides an alternative to current cancer screening methods that use tissue biopsies for the confirmation of diagnosis. This paper attempts to determine how far the regulatory, policy, and governance framework provide support to LB implementation into healthcare systems and how the situation can be improved. For that reason, the European Alliance for Personalised Medicine (EAPM) organized series of expert panels including different key stakeholders to identify different steps, challenges, and opportunities that need to be taken to effectively implement LB technology at the country level across Europe. To accomplish a change of patient care with an LB approach, it is required to establish collaboration between multiple stakeholders, including payers, policymakers, the medical and scientific community, and patient organizations, both at the national and international level. Regulators, pharma companies, and payers could have a major impact in their own domain. Linking national efforts to EU efforts and vice versa could help in implementation of LB across Europe, while patients, scientists, physicians, and kit manufacturers can generate a pull by undertaking more research into biomarkers.
Collapse
Affiliation(s)
- Denis Horgan
- European Alliance for Personalised Medicine, 1040 Brussels, Belgium
- Department of Molecular and Cellular Engineering, Jacob Institute of Biotechnology and Bioengineering, Faculty of Engineering and Technology, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India
| | - Tanja Čufer
- Medical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Francesco Gatto
- Department of Oncology-Pathology, Karolinska Institute, 171 64 Stockholm, Sweden
| | - Iwona Lugowska
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute and Oncology Centre (MSCI), 02781 Warsaw, Poland
| | - Donatella Verbanac
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Ante Kovačića 1, 10000 Zagreb, Croatia
| | - Ângela Carvalho
- i3S—nstituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Jonathan A. Lal
- Department of Molecular and Cellular Engineering, Jacob Institute of Biotechnology and Bioengineering, Faculty of Engineering and Technology, Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj 211007, India
- Institute for Public Health Genomics, Department of Genetics and Cell Biology, GROW School of Oncology and Developmental Biology, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands
| | - Marta Kozaric
- European Alliance for Personalised Medicine, 1040 Brussels, Belgium
| | - Sinead Toomey
- Department of Molecular Medicine, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Smurfit Building, D09 Dublin, Ireland
| | - Hristo Y. Ivanov
- Department of Paediatric and Medical Genetics, Medical University, 4000 Plovdiv, Bulgaria
| | - John Longshore
- Astra Zeneca, 1800 Concord Pike, Wilmington, DE 19803, USA
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, 80137 Naples, Italy
| | - Samantha Hasenleithner
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, 8036 Graz, Austria
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, FHU OncoAge, Pasteur Hospital, University Côte d’Azur, CEDEX 01, 06001 Nice, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, 641 Avenue du Doyen Gaston Giraud, CEDEX 5, 34093 Montpellier, France
| |
Collapse
|
42
|
Potter AJ, Colebatch AJ, Rawson RV, Ferguson PM, Cooper WA, Gupta R, O'Toole S, Saw RPM, Ch'ng S, Menzies AM, Long GV, Scolyer RA. Pathologist initiated reflex BRAF mutation testing in metastatic melanoma: experience at a specialist melanoma treatment centre. Pathology 2022; 54:526-532. [PMID: 35249747 DOI: 10.1016/j.pathol.2021.12.290] [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: 09/23/2021] [Revised: 11/22/2021] [Accepted: 12/02/2021] [Indexed: 11/28/2022]
Abstract
Testing for BRAF mutations in metastatic melanoma is pivotal to identifying patients suitable for targeted therapy and influences treatment decisions regarding single agent versus combination immunotherapy. Knowledge of BRAF V600E immunohistochemistry (IHC) results can streamline decisions during initial oncology consultations, prior to DNA-based test results. In the absence of formal guidelines that require pathologist initiated ('reflex') BRAF mutation testing, our institution developed a local protocol to perform BRAF V600E IHC on specimens from all stage III/IV melanoma patients when the status is otherwise unknown. This study was designed to evaluate the application of this protocol in a tertiary referral pathology department. A total of 408 stage III/IV melanoma patients had tissue specimens accessioned between 1 January and 31 March in three consecutive years (from 2019 to 2021), reported by 32 individual pathologists. The BRAF mutation status was established by pathologists in 87% (352/408) of cases. When a prior BRAF mutation status was previously known, as confirmed in linked electronic records (202/408), this status had been communicated by the clinician on the pathology request form in 1% of cases (3/202). Pathologists performed BRAF V600E IHC in 153 cases (74% of cases where the status was unknown, 153/206) and testing was duplicated in 5% of cases (20/408). Reflex BRAF IHC testing was omitted in 26% of cases (53/206), often on specimens with small volume disease (cytology specimens or sentinel node biopsies) despite adequate tissue for testing. Incorporating BRAF IHC testing within routine diagnostic protocols of stage III/IV melanoma was both feasible and successful in most cases. Communication of a patient's BRAF mutation status via the pathology request form will likely improve implementation of pathologist initiated BRAF mutation testing and may result in a reduction of duplicate tests. To improve pathologist reflex testing rates, we advocate for the use of an algorithmic approach to pathologist initiated BRAF mutation testing utilising both IHC and DNA-based methodologies for stage III/IV melanoma patients.
Collapse
Affiliation(s)
- Alison J Potter
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Andrew J Colebatch
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia
| | - Robert V Rawson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Peter M Ferguson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Wendy A Cooper
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Western Sydney University, Campbelltown, NSW, Australia
| | - Ruta Gupta
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Sandra O'Toole
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Western Sydney University, Campbelltown, NSW, Australia
| | - Robyn P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia; Mater Hospital, North Sydney, NSW, Australia
| | - Sydney Ch'ng
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia; Mater Hospital, North Sydney, NSW, Australia; Chris O'Brien Lifehouse, Camperdown, NSW, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Mater Hospital, North Sydney, NSW, Australia; Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; Mater Hospital, North Sydney, NSW, Australia; Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
43
|
Frisone D, Sandoval J, Friedlaender A, Olivier T, Addeo A. Trends in incidence and mortality of lung cancer in Switzerland: Possible explanations and open questions. Cancer Epidemiol 2022; 80:102232. [PMID: 35905519 DOI: 10.1016/j.canep.2022.102232] [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: 03/23/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Using US population-level data, it has been suggested that novel treatment advances, particularly targeted therapies, have contributed to a sharp fall in NSCLC mortality. Switzerland is a high-income country, with a universal, highly performant health care system, easy access to novel drugs but with different dynamics concerning the smoking epidemic than the US. METHODS We use population-based data from Swiss cancer registries to analyze the trends in incidence, mortality and survival and relate them to recent drug approvals. RESULTS The incidence of NSCLC and SCLC was stable from 1980 to 2018. We noted an important difference between sexes, with an important decrease in men and increase in women, especially for NSCLC. 1-y and 5-y survival have improved for NSCLC between 2004 and 2008 and 2014-2018. CONCLUSION These findings should be regarded as the results of a multifactorial improvement in care and it is difficult for us to pinpoint a unique cause explaining the reduction in mortality.
Collapse
Affiliation(s)
- Daniele Frisone
- Department of Oncology, Geneva University Hospital, SCCL, Switzerland
| | - Jose Sandoval
- Department of Oncology, Geneva University Hospital, SCCL, Switzerland
| | - Alex Friedlaender
- Department of Oncology, Geneva University Hospital, SCCL, Switzerland; Clinique Générale Beaulieu, Geneva, Switzerland
| | - Timothée Olivier
- Department of Oncology, Geneva University Hospital, SCCL, Switzerland; Department of Epidemiology and Biostatistics, University of California San Francisco, 550 16th St, 2nd Fl, San Francisco, CA 94158, USA
| | - Alfredo Addeo
- Department of Oncology, Geneva University Hospital, SCCL, Switzerland; Geneva University, Faculty of Medicine, Switzerland.
| |
Collapse
|
44
|
Davies M. Oncogenic-Directed Therapy for Advanced Non-Small Cell Lung Cancer: Implications for the Advanced Practice Nurse. Clin J Oncol Nurs 2022; 26:245-251. [PMID: 35604726 DOI: 10.1188/22.cjon.245-251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Molecular profiling and testing for oncogenic driver mutations is an essential component in the diagnosis of patients with advanced non-small cell lung cancer (NSCLC). Results of these tests guide personalized targeted therapy in patients with NSCLC harboring an oncogenic driver. Advanced practice nurses are at the center of coordinating care for patients with NSCLC from the time of diagnosis and have a role in assuring appropriate testing is ordered and therapy is selected based on testing results.
Collapse
|
45
|
McCoach CE, Rolfo C, Drilon A, Lacouture M, Besse B, Goto K, Zhu VW, Tan DSW, Farajian S, Potter LA, Kherani JF, Soldatenkova V, Olek EA, Muehlenbein CE, Park K. Hypersensitivity Reactions to Selpercatinib Treatment With or Without Prior Immune Checkpoint Inhibitor Therapy in Patients With NSCLC in LIBRETTO-001. J Thorac Oncol 2022; 17:768-778. [PMID: 35183775 PMCID: PMC11083849 DOI: 10.1016/j.jtho.2022.02.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 02/01/2022] [Accepted: 02/07/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Immune checkpoint inhibitor (ICI) therapy has been found to increase the risk/severity of immune-mediated adverse events with subsequent kinase inhibitor treatment in oncogenically driven cancers. We explored the risk for hypersensitivity with selpercatinib, a first-in-class highly selective and potent, central nervous system-active RET inhibitor, in prior ICI-treated patients with RET fusion-positive NSCLC compared with their ICI-naive counterparts. METHODS Data from patients enrolled by December 16, 2019, in the ongoing phase 1/2 LIBRETTO-001 (NCT03157128) trial were analyzed for hypersensitivity reactions reported using preferred terms of hypersensitivity/drug hypersensitivity and defined as a constellation of symptoms/findings characterized by maculopapular rash, often preceded by fever with arthralgias/myalgias, followed by greater than or equal to 1 of the following signs/symptoms: thrombocytopenia, increased aspartate aminotransferase or alanine aminotransferase, hypotension, tachycardia, or increased creatinine. RESULTS Of 329 patients, 22 (7%) who experienced a grade 1 to 3 hypersensitivity reaction that met the defined constellation of events were attributed to selpercatinib by investigators, and more often in prior ICI-treated (n = 17, 77%) than ICI-naive (n = 5, 23%) patients. There were 19 patients with selpercatinib-related hypersensitivity who resumed selpercatinib post-hypersensitivity with dose modification/supportive care. Furthermore, 17 patients, of whom 14 received prior ICI therapy, were still on treatment at twice daily doses of 40 mg (n = 5), 80 mg (n = 4), 120 mg (n = 4), and 160 mg (n = 4). CONCLUSIONS Rates of selpercatinib-related hypersensitivity were low overall and, as with other kinase inhibitors, occurred predominantly in prior ICI-treated patients. Hypersensitivity to selpercatinib can be managed with supportive care measures regardless of prior ICI status and is reversible.
Collapse
Affiliation(s)
- Caroline E McCoach
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California; Present Address: Genentech, Inc., South San Francisco, California.
| | - Christian Rolfo
- Greenebaum Comprehensive Cancer Center, Experimental Therapeutics Program, School of Medicine, University of Maryland, Baltimore, Maryland; Present Address: Center for Thoracic Oncology at Tisch Cancer Institute, Mount Sinai Health System and Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Mario Lacouture
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Koichi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Viola W Zhu
- Department of Medicine, Division of Hematology and Oncology, University of California Irvine, Orange, California
| | - Daniel S W Tan
- National Cancer Centre Singapore, Duke-National University of Singapore Medical School, Singapore
| | - Stephanie Farajian
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Laura A Potter
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California; Present Address: Davis School of Medicine, University of California, Sacramento, Sacramento, California
| | - Jennifer F Kherani
- Loxo Oncology, Inc., a wholly owned subsidiary of Eli Lilly and Company, Stamford, Connecticut
| | | | - Elizabeth A Olek
- Loxo Oncology, Inc., a wholly owned subsidiary of Eli Lilly and Company, Stamford, Connecticut
| | | | - Keunchil Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
46
|
Wang H, Zhou X, Wang Z, Lu T, Li B, Jiang S. Clinical Efficacy of Osimertinib in Patients with Advanced Non-Small Cell Lung Cancer and Its Effect on Serum CEA and VEGF Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:3032087. [PMID: 35664943 PMCID: PMC9159882 DOI: 10.1155/2022/3032087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/22/2022] [Accepted: 04/26/2022] [Indexed: 11/17/2022]
Abstract
Objective To assess the clinical efficacy of osimertinib in patients with advanced non-small cell lung cancer and its effect on serum carcinoembryonic antigen (CEA) and vascular endothelial growth factor (VEGF) expression. Methods Between July 2018 and January 2020, 80 patients with advanced non-small cell lung cancer were assessed for eligibility and recruited. The patients were assigned at a ratio of 1 : 1 to receive either the PC regimen (pemetrexed + cisplatin) (conventional group) or osimertinib (experimental group). The primary endpoint was the clinical efficacy, and the secondary endpoints were the adverse events, expression of serum CEA and VEGF, and 2-year survival. Results Osimertinib was associated with a significantly higher response rate and disease control rate versus pemetrexed plus cisplatin (P < 0.05). Osimertinib resulted in a significantly lower incidence of adverse events versus the PC regimen (P < 0.05). Patients given osimertinib had significantly lower levels of CEA and VEGF versus those given pemetrexed plus cisplatin (P < 0.05). Osimertinib was associated with a significantly higher 1-year and 2-year survival rate versus pemetrexed plus cisplatin. Conclusion Osimertinib could inhibit the expression of serum CEA and VEGF in patients with advanced non-small cell lung cancer and reduce the adverse events with significant efficacy, so it is worthy of clinical promotion and application.
Collapse
Affiliation(s)
- Huanyuan Wang
- Department of Thoracic Surgery, Jiangxi Cancer Hospital of Nanchang University, Nanchang 330029, China
| | - Xiangwu Zhou
- Department of Thoracic Surgery, Medical College of Nanchang University, Nanchang 330006, China
| | - Zhaozhen Wang
- Department of Clinical Medicine, Jiangxi Health Vocational College of China, Nanchang, Jiangxi, China
| | - Tianzhu Lu
- Department of Radiation Oncology, Jiangxi Cancer Hospital of Nanchang University, Nanchang 330029, China
| | - Baoliang Li
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopaedic Research Institute/Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Sicong Jiang
- Division of Thoracic and Endocrine Surgery, University Hospitals and University of Geneva, Geneva 1211, Geneva 4, Switzerland
| |
Collapse
|
47
|
Use of the Biocartis Idylla™ Platform for the Detection of Epidermal Growth Factor Receptor, BRAF and KRAS Proto-Oncogene Mutations in Liquid-Based Cytology Specimens from Patients with Non-Small Cell Lung Carcinoma and Pancreatic Adenocarcinoma. JOURNAL OF MOLECULAR PATHOLOGY 2022. [DOI: 10.3390/jmp3020010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The study aimed to demonstrate rapid and effective molecular testing on liquid-based cytology (LBC) samples for EGFR, KRAS and BRAF mutations using the Biocartis Idylla™. Rapid on-site evaluation (ROSE) LBC samples for patients with non-small cell lung carcinoma (NSCLC) or pancreatic ductal adenocarcinoma (PDAC) were tested for EGFR, KRAS and BRAF mutations based on the relevance to tumour subtype. The quantification values (Cq values) and mutation detection status were compared between LBC samples and routine formalin-fixed paraffin-embedded (FFPE) clot samples. ROSE LBC samples (n = 54) showed a higher yield of well-preserved tumour and wild type (WT) DNA, demonstrated by lower quantification cycles, no false positives or false negatives, and a higher sensitivity for low allele frequency mutations when compared with FFPE clot samples. The Biocartis Idylla™ provides highly sensitive, reliable and rapid testing for LBC samples for the detection of EFGR and KRAS mutations. BRAF mutations were not detected in the participant cohort; however, all LBC WT BRAF results correlated with the results from the FFPE clot samples. Access to rapid molecular testing using LBC samples can detect the most frequent driver mutations closer to the time of diagnosis, enabling the selection of the most effective first-line targeted therapy sooner, reducing delays or side effects from suboptimal treatments, patient anxiety and costs to healthcare systems, whilst improving patient outcomes.
Collapse
|
48
|
Klarenbeek SE, Aarts MJ, van den Heuvel MM, Prokop M, Tummers M, Schuurbiers OCJ. Impact of time-to-treatment on survival for advanced non-small cell lung cancer patients in the Netherlands: a nationwide observational cohort study. Thorax 2022; 78:467-475. [PMID: 35450944 DOI: 10.1136/thoraxjnl-2021-218059] [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: 08/05/2021] [Accepted: 03/21/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND The assumption that more rapid treatment improves survival of advanced non-small cell lung cancer (NSCLC) has not yet been proven. We studied the relation between time-to-treatment and survival in advanced stage NSCLC patients in a large multicentric nationwide retrospective cohort. Additionally, we identified factors associated with delay. METHOD We selected 10 306 patients, diagnosed and treated between 2014 and 2019 for clinical stage III and IV NSCLC, from the Netherlands Cancer Registry that includes nationwide data from 109 Dutch hospitals. Associations between survival and time-to-treatment were tested with Cox proportional hazard regression analyses. Time-to-treatment was adjusted for multiple covariates including diagnostic procedures and type of therapy. Factors associated with delay were identified by multilevel logistic regression. RESULTS Risk of death significantly decreased with longer time-to-treatment for stage III patients receiving only radiotherapy (adjusted HR, aHR >21 days: 0.59 (95% CI 0.48 to 0.73)) or any type of systemic therapy (aHR >49 days: 0.72 (95% CI 0.56 to 0.91)) and stage IV patients receiving chemotherapy and/or immunotherapy (aHR >21 days: 0.81 (95% CI 0.73 to 0.88)). No significant association was found for stage III patients treated with chemoradiotherapy and stage IV patients treated with targeted therapy. More complex diagnostic procedures often delay treatment. CONCLUSION Although in general it is important to start treatment as early as possible, our study finds no evidence that a more rapid start of treatment improves outcomes in advanced stage NSCLC patients. The benefit of urgent treatment is probably confounded by unmeasured patient and tumour characteristics and, clinical urgency dictating timelines of treatment. Time-to-treatment and its impact should be continuously evaluated as therapeutic strategies continue to evolve and improve.
Collapse
Affiliation(s)
- Sosse E Klarenbeek
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mieke J Aarts
- Research and Development, Dutch Association of Comprehensive Cancer Centres, Utrecht, The Netherlands
| | - Michel M van den Heuvel
- Department of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mathias Prokop
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcia Tummers
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Olga C J Schuurbiers
- Department of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
49
|
Liao X, Liu M, Wang R, Zhang J. Potentials of Non-Invasive 18F-FDG PET/CT in Immunotherapy Prediction for Non-Small Cell Lung Cancer. Front Genet 2022; 12:810011. [PMID: 35186013 PMCID: PMC8855498 DOI: 10.3389/fgene.2021.810011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/31/2021] [Indexed: 12/26/2022] Open
Abstract
The immune checkpoint inhibitors (ICIs), by targeting cytotoxic-T-lymphocyte-associated protein 4, programmed cell death 1 (PD-1), or PD-ligand 1, have dramatically changed the natural history of several cancers, including non-small cell lung cancer (NSCLC). There are unusual response manifestations (such as pseudo-progression, hyper-progression, and immune-related adverse events) observed in patients with ICIs because of the unique mechanisms of these agents. These specific situations challenge response and prognostic assessment to ICIs challenging. This review demonstrates how 18F-FDG PET/CT can help identify these unusual response patterns in a non-invasive and effective way. Then, a series of semi-quantitative parameters derived from 18F-FDG PET/CT are introduced. These indexes have been recognized as the non-invasive biomarkers to predicting the efficacy of ICIs and survival of NSCLC patients according to the latest clinical studies. Moreover, the current situation regarding the functional criteria based on 18F-FDG PET/CT for immunotherapeutic response assessment is presented and analyzed. Although the criteria based on 18F-FDG PET/CT proposed some resolutions to overcome limitations of morphologic criteria in the assessment of tumor response to ICIs, further researches should be performed to validate and improve these assessing systems. Then, the last part in this review displays the present status and a perspective of novel specific PET probes targeting key molecules relevant to immunotherapy in prediction and response assessment.
Collapse
Affiliation(s)
| | | | | | - Jianhua Zhang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| |
Collapse
|
50
|
Schneider MA, Rozy A, Wrenger S, Christopoulos P, Muley T, Thomas M, Meister M, Welte T, Chorostowska-Wynimko J, Janciauskiene S. Acute Phase Proteins as Early Predictors for Immunotherapy Response in Advanced NSCLC: An Explorative Study. Front Oncol 2022; 12:772076. [PMID: 35174082 PMCID: PMC8841510 DOI: 10.3389/fonc.2022.772076] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/10/2022] [Indexed: 01/22/2023] Open
Abstract
In the last decade, targeting the immune system became a promising therapy in advanced lung cancer stages. However, in a clinical follow-up, patient responses to immune checkpoint inhibitors widely differ. Peripheral blood is a minimally invasive source of potential biomarkers to explain these differences. We blindly analyzed serum samples from 139 patients with non-small cell lung cancer prior to anti-PD-1 or anti-PD-L1 therapies to assess whether baseline levels of albumin (ALB), alpha-1 acid glycoprotein (AGP), alpha1-antitrypsin (AAT), alpha2-macroglobulin (A2M), ceruloplasmin (CP), haptoglobin (HP), alpha1-antichymotrypsin (ACT), serum amyloid A (SAA), and high-sensitivity C-reactive protein (hs-CRP), have a predictive value for immunotherapy success. Disease progression-free survival (PFS) was calculated based on RECIST 1.1 criteria. A multivariate Cox regression analysis, including serum levels of acute-phase proteins and clinical parameters, revealed that higher pre-therapeutic levels of HP and CP are independent predictors of a worse PFS. Moreover, a combined panel of HP and CP stratified patients into subgroups. We propose to test this panel as a putative biomarker for assessing the success of immunotherapy in patients with NSCLC.
Collapse
Affiliation(s)
- Marc A. Schneider
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
- Translational Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Adriana Rozy
- Laboratory of Molecular Diagnostics and Immunology, National Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
| | - Sabine Wrenger
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- Biomedical Research in End Stage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Petros Christopoulos
- Translational Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Thoracic Oncology, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Muley
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
- Translational Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Michael Thomas
- Translational Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Thoracic Oncology, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Meister
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
- Translational Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- Biomedical Research in End Stage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Joanna Chorostowska-Wynimko
- Laboratory of Molecular Diagnostics and Immunology, National Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
| | - Sabina Janciauskiene
- Laboratory of Molecular Diagnostics and Immunology, National Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- Biomedical Research in End Stage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
- *Correspondence: Sabina Janciauskiene,
| |
Collapse
|