151
|
Gadgeel SM, Rodríguez-Abreu D, Halmos B, Garassino MC, Kurata T, Cheng Y, Jensen E, Shamoun M, Rajagopalan K, Paz-Ares L. Pembrolizumab Plus Chemotherapy for Metastatic NSCLC With Programmed Cell Death Ligand 1 Tumor Proportion Score Less Than 1%: Pooled Analysis of Outcomes After Five Years of Follow-Up. J Thorac Oncol 2024; 19:1228-1241. [PMID: 38642841 DOI: 10.1016/j.jtho.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/11/2024] [Accepted: 04/14/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND We report long-term outcomes from a pooled analysis of patients with previously untreated metastatic NSCLC with programmed cell death ligand 1 (PD-L1) tumor proportion score (TPS) less than 1% enrolled in phase III studies of pembrolizumab plus chemotherapy versus placebo plus chemotherapy. METHODS This exploratory pooled analysis included individual patient data from the KEYNOTE-189 global (NCT02578680) and Japan extension (NCT03950674) studies of metastatic nonsquamous NSCLC without EGFR or ALK alterations and the KEYNOTE-407 global (NCT02775435) and People's Republic of China extension (NCT03875092) studies of metastatic squamous NSCLC. Patients received pembrolizumab or placebo plus pemetrexed and cisplatin or carboplatin in KEYNOTE-189 and pembrolizumab or placebo plus carboplatin and paclitaxel or nab-paclitaxel in KEYNOTE-407. PD-L1 TPS was centrally assessed using PD-L1 IHC 22C3 pharmDx (Agilent Technologies, Carpinteria, CA). RESULTS Overall, 442 patients were included in this analysis (pembrolizumab plus chemotherapy, n = 255; chemotherapy, n = 187). The median follow-up was 60.7 (range, 49.9‒72.0) months. Pembrolizumab plus chemotherapy improved overall survival (hazard ratio, 0.64; 95% confidence interval [CI]: 0.51‒0.79) and progression-free survival (hazard ratio, 0.66; 95% CI: 0.54‒0.81) versus chemotherapy. The 5-year overall survival rates (95% CI) were 12.5% (8.6%‒17.3%) versus 9.3% (5.6%‒14.1%). Grades 3 to 5 treatment-related adverse events occurred in 59.1% of patients for pembrolizumab plus chemotherapy and 61.3% for chemotherapy. CONCLUSION With approximately 5 years of follow-up, pembrolizumab plus chemotherapy provided clinically meaningful and durable improvements in survival outcomes versus chemotherapy alone in patients with previously untreated metastatic NSCLC with PD-L1 TPS less than 1%. These results continue to support pembrolizumab plus chemotherapy as a standard of care in this patient population. CLINICALTRIALS gov, NCT02578680 (KEYNOTE-189 global), NCT03950674 (KEYNOTE-189 Japan extension), NCT02775435 (KEYNOTE-407 global), NCT03875092 (KEYNOTE-407 People's Republic of China extension).
Collapse
Affiliation(s)
| | - Delvys Rodríguez-Abreu
- Complejo Hospitalario Universitario Insular Materno-Infantil de Gran Canaria, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Balazs Halmos
- Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York
| | - Marina C Garassino
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Current Affiliation: Thoracic Oncology Program, The University of Chicago Medicine & Biological Sciences, Chicago, Illinois
| | - Takayasu Kurata
- Department of Thoracic Oncology, Kansai Medical University Hospital, Osaka, Japan
| | - Ying Cheng
- Department of Oncology, Jilin Cancer Hospital, Changchun, People's Republic of China
| | | | | | | | - Luis Paz-Ares
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Universidad Complutense and Ciberonc, Madrid, Spain
| |
Collapse
|
152
|
Park J, Lee B, Song JY, Sung M, Kwon MJ, Kim CR, Lee S, Shin YK, Choi YL. Detection of EGFR exon 20 insertion mutations in non-small cell lung cancer: implications for consistent nomenclature in precision medicine. Pathology 2024; 56:653-661. [PMID: 38811262 DOI: 10.1016/j.pathol.2024.02.012] [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: 10/06/2023] [Revised: 01/11/2024] [Accepted: 02/23/2024] [Indexed: 05/31/2024]
Abstract
Epidermal growth factor receptor (EGFR) exon 20 insertion mutations (E20ins) are the third most frequent mutations observed in non-small cell lung cancer, accounting for approximately 1-10% of all EGFR mutations. In the era of precision medicine and targeted therapies, consistent naming of genetic alterations is crucial to avoid confusion and errors. However, the annotation of EGFR E20ins mutations has been inconsistent, leading to confusion in the scientific literature and product documentation. In this study, our primary objective was to investigate the usage of different annotation related to EGFR E20ins in independent studies. Additionally, we assessed the distribution of EGFR E20ins mutations and estimated the detection coverage expected from each available EGFR E20ins detection assay. A total of 1,418 EGFR E20ins mutations were collected from six studies (FoundationInsights, Geneseeq Technology Inc, mobocertinib phase I/II trial, poziotinib phase II trial, sunvozertinib phase I trial, and Samsung Medical Center) and reorganised according to Human Genome Variation Society (HGVS) nomenclature. Our analysis revealed that the majority of EGFR E20ins mutations requiring correction were 'insertion' or 'deletion-insertion', which should be appropriately designated as 'duplication'. Additionally, duplicated variants were reported using different annotations in each study, and furthermore, even identical variant sequences were annotated differently within the same study. In all six studies, p.A767_V769dup and p.S768_D770dup were the most frequently observed EGFR E20ins. The Oncomine Dx Target Test showed the highest patient coverage at 77.2%, followed by the Droplex EGFR Mutation Test v2 with a patient coverage of 70.5% for EGFR E20ins patients. To ensure comprehensive coverage in real-world settings, it is essential to standardise the annotations for each variant, for example using the HGVS nomenclature. The accurate classification and analysis of drug responsiveness in EGFR E20ins necessitate consideration of the nomenclature, particularly with respect to the locations where the actual mutations occur.
Collapse
Affiliation(s)
- Jieun Park
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Boram Lee
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ji-Young Song
- Laboratory of Molecular Pathology and Theranostics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Minjung Sung
- Laboratory of Molecular Pathology and Theranostics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Mi Jeong Kwon
- Vessel-Organ Interaction Research Center (MRC), College of Pharmacy, Kyungpook National University, Daegu, South Korea; BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Chae Rin Kim
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Sangjin Lee
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Young Kee Shin
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea; Laboratory of Molecular Pathology and Cancer Genomics, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; Laboratory of Molecular Pathology and Theranostics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, South Korea.
| |
Collapse
|
153
|
Cheung WY, Carbonell C, Navani V, Sangha RS, Ewara EM, Elia-Pacitti J, Iczkovitz S, Jarada TN, Warkentin MT. Real-World Treatment Patterns, Health Outcomes, and Healthcare Resource Use in Advanced Common EGFR-Positive Non-Small Cell Lung Cancer Patients Treated with Osimertinib in Alberta. Curr Oncol 2024; 31:4382-4396. [PMID: 39195310 PMCID: PMC11352775 DOI: 10.3390/curroncol31080327] [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: 05/28/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
There is limited information on the treatment trajectory and outcomes of patients with advanced cEGFRm NSCLC treated with osimertinib in routine clinical practice in Canada. By using and analyzing population-based administrative data and detailed chart abstraction in the province of Alberta, our objective was to capture Canadian-specific real-world treatment patterns, health outcomes, and healthcare resource utilization (HCRU) in advanced cEGFRm NSCLC patients who were (a) treated with osimertinib and (b) those receiving treatment after osimertinib. In our study cohort, we found that the overall survival rates for real-world patients receiving osimertinib were less favorable than those observed in clinical trials (24.0 versus 38.6 months). The attrition rate after osimertinib was substantial and high HCRU persisted across many years after diagnosis and treatment. This study provides important real-world evidence on contemporary survival, treatment patterns, and healthcare use among cEGFRm NSCLC patients treated with osimertinib and suggests that further research efforts are needed to improve therapeutic options in both the first and subsequent line settings.
Collapse
Affiliation(s)
- Winson Y. Cheung
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada (T.N.J.)
| | - Chantelle Carbonell
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada (T.N.J.)
| | - Vishal Navani
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada (T.N.J.)
| | - Randeep S. Sangha
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | | | | | - Sandra Iczkovitz
- Johnson & Johnson Innovative Medicine, Toronto, ON M3C 1L9, Canada
| | - Tamer N. Jarada
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada (T.N.J.)
| | - Matthew T. Warkentin
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada (T.N.J.)
| |
Collapse
|
154
|
Peters HA, Weiss D, Boschheidgen M, Mamlins E, Giesel FL, Fluegen G, Kirchner J, Antoch G, Jannusch K. Prognostic potential of integrated morphologic and metabolic parameters of pre-therapeutic [18F]FDG-PET/CT regarding progression-free survival (PFS) and overall survival (OS) in NSCLC-patients. PLoS One 2024; 19:e0307998. [PMID: 39074093 DOI: 10.1371/journal.pone.0307998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024] Open
Abstract
PURPOSE This study aimed to evaluate the prognostic potential of pre-therapeutic [18F]FDG-PET/CT variables regarding prediction of progression-free survival (PFS) and overall survival (OS) in NSCLC-patients. METHOD NSCLC-patients who underwent pre-therapeutic [18F]FDG-PET/CT were retrospectively analyzed. The following imaging features were collected from the primary tumor: tumor size, tumor density, central necrosis, spicules and SUVmax. For standardization, an indexSUVmax was calculated (SUVmax primary tumor/SUVmax liver). Descriptive statistics and correlations of survival time analyses for PFS and OS were calculated using the Kaplan-Meier method and Cox regression including a hazard ratio (HR). A value of p < 0.05 was set as statistically significant. The 95%-confidence intervals (CI) were calculated. The median follow-up time was 63 (IQR 27-106) months. RESULTS This study included a total of 82 patients (25 women, 57 men; mean age: 66 ± 9 years). IndexSUVmax (PFS: HR = 1.0, CI: 1.0-1.1, p = 0.49; OS: HR = 1.0, CI: 0.9-1.2, p = 0.41), tumor size (PFS: HR = 1.0, CI: 0.9-1.0, p = 0.08; OS: HR = 1.0, CI: 0.9-1.0, p = 0.07), tumor density (PFS: HR = 0.9, CI: 0.6-1.4, p = 0.73; OS: HR = 0.3; CI: 0.1-1.1; p = 0.07), central necrosis (PFS: HR = 1.0, CI: 0.6-1.8, p = 0.98; OS: HR = 0.6, CI: 0.2-1.9, p = 0.40) and spicules (PFS: HR = 1.0, CI: 0.6-1.9, p = 0.91; OS: HR = 1.3, CI: 0.4-3.7, p = 0.65) did not significantly affect PFS and OS in the study population. An optimal threshold value for the indexSUVmax was determined by ROC analysis and Youden's index. There was no significant difference in PFS with an indexSUVmax-threshold of 3.8 (13 vs. 27 months; p = 0.45) and in OS with an indexSUVmax-threshold of 4.0 (113 vs. 106 months; p = 0.40). CONCLUSIONS SUVmax and morphologic parameters from pre-therapeutic [18F]FDG-PET/CT were not able to predict PFS and OS in NSCLC-patients.
Collapse
Affiliation(s)
- Helena A Peters
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Daniel Weiss
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Matthias Boschheidgen
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Eduards Mamlins
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Frederik L Giesel
- Department of Nuclear Medicine, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Georg Fluegen
- Department of Surgery, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Julian Kirchner
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Gerald Antoch
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Kai Jannusch
- Department of Diagnostic and Interventional Radiology, Medical Faculty and University Hospital Duesseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
155
|
Oswalt CJ, Nakatani MM, Troy J, Wolf S, Locke SC, LeBlanc TW. Timing of Palliative Care Consultation Impacts End of Life Care Outcomes in Metastatic Non-Small Cell Lung Cancer. J Pain Symptom Manage 2024:S0885-3924(24)00858-3. [PMID: 39002711 DOI: 10.1016/j.jpainsymman.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/02/2024] [Accepted: 07/07/2024] [Indexed: 07/15/2024]
Abstract
CONTEXT Early specialist palliative care (PC) involvement in metastatic non-small cell lung cancer (mNSCLC) is associated with improved quality of life, less aggressive end of life (EoL) care, and longer survival. As treatment paradigms for NSCLC have evolved, PC utilization remains low. OBJECTIVES This work examines how the timing and extent of PC involvement impacts outcomes and the patient experience in mNSCLC in the era of immunotherapy. METHODS This retrospective review analyzed patients with mNSCLC who initiated first-line treatment with chemotherapy, immunotherapy, or combined chemoimmunotherapy at Duke University between March 2015 and July 2019. PC consultation and outcomes data were abstracted through November 2022. EoL care variables were analyzed using descriptive statistics. RESULTS 152 patients were stratified based on whether PC was consulted during their disease course. 80 patients (53%) never saw PC, while the 72 patients (47%) who saw PC were further stratified by time to first PC encounter and total number of PC visits. 31% were seen within two months of diagnosis (early), 33% between two and six months (intermediate), and 36% after 6 months (late). Patients who received early PC had longer median time on hospice (35 days), had lower rates of aggressive EoL care (43%), and experienced less frequent in-hospital death (14%) compared to other groups. CONCLUSION This real-world study reveals that referrals to PC still occur late or not at all in mNSCLC despite demonstrated benefits of early PC integration. Early outpatient PC referrals resulted in longer time on hospice, lower frequency of aggressive EoL care, and lower rates of in-hospital death.
Collapse
Affiliation(s)
- Cameron J Oswalt
- Duke Cancer Institute (C.J.O., S.C.L., T.W.L.B.,), Durham, North Carolina, USA.
| | - Morgan M Nakatani
- Medicine-Psychiatry Resident (M.M.N.), Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Jesse Troy
- Department of Biostatistics and Bioinformatics (J.T., S.W.), Division of Biostatistics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Steven Wolf
- Department of Biostatistics and Bioinformatics (J.T., S.W.), Division of Biostatistics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Susan C Locke
- Duke Cancer Institute (C.J.O., S.C.L., T.W.L.B.,), Durham, North Carolina, USA
| | - Thomas W LeBlanc
- Duke Cancer Institute (C.J.O., S.C.L., T.W.L.B.,), Durham, North Carolina, USA
| |
Collapse
|
156
|
Hochmair M, Terbuch A, Lang D, Trockenbacher C, Augustin F, Ghanim B, Maurer D, Taghizadeh H, Kamhuber C, Wurm R, Lindenmann J, Braz P, Bundalo T, Begic M, Bauer J, Reimann P, Müser N, Huemer F, Schlintl V, Bianconi D, Baumgartner B, Schenk P, Rauter M, Hötzenecker K. Real-World Treatment Patterns and Timeliness of Clinical Care Pathway for Non-Small Cell Lung Cancer Patients in Austria: The PRATER Retrospective Study. Cancers (Basel) 2024; 16:2586. [PMID: 39061224 PMCID: PMC11275022 DOI: 10.3390/cancers16142586] [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: 06/11/2024] [Revised: 07/05/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
This was a retrospective study of the profile and initial treatments of adults diagnosed with early-stage (ES) non-small cell lung cancer (NSCLC) during January 2018-December 2021 at 16 leading hospital institutions in Austria, excluding patients enrolled in clinical trials. In total, 319 patients were enrolled at a planned ~1:1:1 ratio across StI:II:III. Most tested biomarkers were programmed death ligand 1 (PD-L1; 58% expressing), Kirsten rat sarcoma virus (KRAS; 22% positive), and epidermal growth factor receptor (EGFR; 18% positive). Of 115/98/106 StI/II/III patients, 82%/85%/36% underwent surgery, followed by systemic therapy in 9%/45%/47% of those [mostly chemotherapy (ChT)]. Unresected treated StIII patients received ChT + radiotherapy [43%; followed by immune checkpoint inhibitors (ICIs) in 39% of those], ICI ± ChT (35%), and ChT-alone/radiotherapy-alone (22%). Treatment was initiated a median (interquartile range) of 24 (7-39) days after histological confirmation, and 55 (38-81) days after first medical visit. Based on exploratory analyses of all patients newly diagnosed with any stage NSCLC during 2018-2021 at 14 of the sites (N = 7846), 22%/10%/25%/43% had StI/II/III/IV. The total number was not significantly different between pre-COVID-19 (2018-2019) and study-specific COVID-19 (2020-2021) periods, while StI proportion increased (21% vs. 23%; p = 0.012). Small differences were noted in treatments. In conclusion, treatments were aligned with guideline recommendations at a time which preceded the era of ICIs and targeted therapies in the (neo)adjuvant setting.
Collapse
Affiliation(s)
- Maximilian Hochmair
- Department of Respiratory and Critical Care Medicine, Karl Landsteiner Institute of Lung Research and Pulmonary Oncology, Klinik Floridsdorf, 1210 Vienna, Austria
| | - Angelika Terbuch
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | - David Lang
- Department of Pulmonology, Johannes Kepler University Linz, Kepler University Hospital, 4829 Linz, Austria
| | | | - Florian Augustin
- Department of Visceral, Transplant and Thoracic Surgery, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Bahil Ghanim
- Department of General and Thoracic Surgery, University Hospital Krems, 3500 Krems an der Donau, Austria
| | - Dominik Maurer
- Department of Pulmonology, Ordensklinikum Elisabethinen Linz, 4020 Linz, Austria
| | - Hossein Taghizadeh
- Division of Oncology, Department of Internal Medicine I, University Hospital St. Pölten, 3100 St. Pölten, Austria
| | - Christoph Kamhuber
- Department of Oncology, Kardinal Schwarzenberg Klinikum, 5620 Schwarzach, Austria
| | - Robert Wurm
- Department of Pulmonology, Medical University Graz, 8036 Graz, Austria
| | - Jörg Lindenmann
- Division of Thoracic and Hyperbaric Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Petra Braz
- Department of Pulmonology, Landesklinikum Hochegg, 2840 Hochegg, Austria (P.S.)
| | - Tatjana Bundalo
- Department of Pulmonology, Landesklinikum Hochegg, 2840 Hochegg, Austria (P.S.)
| | - Merjem Begic
- Department of Thoracic Surgery, Medical University of Vienna, 8036 Vienna, Austria
| | - Johanna Bauer
- Department of Thoracic Surgery, Medical University of Vienna, 8036 Vienna, Austria
| | - Patrick Reimann
- Department of Oncology, Landeskrankenhaus Feldkirch, 6800 Feldkirch, Austria
| | - Nino Müser
- Department of Medicine II with Pneumology, Karl Landsteiner Institute for Lung Research and Pulmonary Oncology, Klinik Ottakring, 1160 Vienna, Austria
| | - Florian Huemer
- Division of Pulmonology, Klinik Penzing, 1140 Vienna, Austria
| | - Verena Schlintl
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria
| | | | | | - Peter Schenk
- Department of Pulmonology, Landesklinikum Hochegg, 2840 Hochegg, Austria (P.S.)
| | - Markus Rauter
- Department of Pulmonology, Klinikum Klagenfurt Am Woerthersee, 9020 Klagenfurt, Austria
| | - Konrad Hötzenecker
- Department of Thoracic Surgery, Medical University of Vienna, 8036 Vienna, Austria
| |
Collapse
|
157
|
Zapata-García M, Moratiel-Pellitero A, Isla D, Gálvez E, Gascón-Ruiz M, Sesma A, Barbero R, Galeano J, del Campo R, Ocáriz M, Quílez E, Cruellas M, Remírez-Labrada A, Pardo J, Martínez-Lostao L, Domingo MP, Esteban P, Torres-Ramón I, Yubero A, Paño JR, Lastra R. Impact of antibiotics, corticosteroids, and microbiota on immunotherapy efficacy in patients with non-small cell lung cancer. Heliyon 2024; 10:e33684. [PMID: 39050456 PMCID: PMC11268177 DOI: 10.1016/j.heliyon.2024.e33684] [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: 02/18/2024] [Revised: 06/10/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Lung cancer is a leading cause of morbidity and mortality globally, with its high mortality rate attributed mainly to non-small cell lung cancer (NSCLC). Although immunotherapy with immune checkpoint inhibitors (ICI) has revolutionized its treatment, patient response is highly variable and lacking predictive markers. We conducted a prospective study on 55 patients with NSCLC undergoing ICI therapy to identify predictive markers of both response and immune-related adverse events (IrAEs) in the airway microbiota. We also analyzed the clinical evolution and overall survival (OS) with respect to treatments that affect the integrity of the microbiota, such as antibiotics and corticosteroids. Our results demonstrated that respiratory microbiota differ significantly in ICI responders: they have higher alpha diversity values and lower abundance of the Firmicutes phylum and the Streptococcus genus. Employing a logistic regression model, the abundance of Gemella was the major predictor of non-ICI response, whereas Lachnoanaerobaculum was the best predictor of a positive response to ICI. The most relevant results were that antibiotic consumption is linked to a lower ICI response, and the use of corticosteroids correlated with poorer overall survival. Whereas previous studies have focused on gut microbiota, our findings highlight the importance of the respiratory microbiota in predicting the treatment response. Future research should explore microbiota modulation strategies to enhance immunotherapy outcomes. Understanding the impact of antibiotics, corticosteroids, and microbiota on NSCLC immunotherapy will help personalize treatment and improve patient outcomes.
Collapse
Affiliation(s)
- María Zapata-García
- Medical Oncology Department, Lozano Blesa University Hospital Clinic, 50009, Zaragoza, Spain
- Health Research Institute of Aragón (IIS Aragón), 50009, Zaragoza, Spain
| | - Alba Moratiel-Pellitero
- Medical Oncology Department, Lozano Blesa University Hospital Clinic, 50009, Zaragoza, Spain
| | - Dolores Isla
- Medical Oncology Department, Lozano Blesa University Hospital Clinic, 50009, Zaragoza, Spain
- Health Research Institute of Aragón (IIS Aragón), 50009, Zaragoza, Spain
| | - Eva Gálvez
- Institute of Carbochemistry (ICB-CSIC), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
| | - Marta Gascón-Ruiz
- Health Research Institute of Aragón (IIS Aragón), 50009, Zaragoza, Spain
- Medical Oncology Department, Miguel Servet University Hospital, 50009, Zaragoza, Spain
| | - Andrea Sesma
- Health Research Institute of Aragón (IIS Aragón), 50009, Zaragoza, Spain
- Medical Oncology Department, Miguel Servet University Hospital, 50009, Zaragoza, Spain
| | - Raquel Barbero
- Microbiology Department, Ramón y Cajal University Hospital and IRYCIS, Madrid, Spain
| | - Javier Galeano
- Complex Systems Group, Universidad Politécnica de Madrid, Madrid, Spain
| | - Rosa del Campo
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
- Microbiology Department, Ramón y Cajal University Hospital and IRYCIS, Madrid, Spain
| | - Maitane Ocáriz
- Medical Oncology Department, Lozano Blesa University Hospital Clinic, 50009, Zaragoza, Spain
| | - Elisa Quílez
- Medical Oncology Department, Lozano Blesa University Hospital Clinic, 50009, Zaragoza, Spain
- Health Research Institute of Aragón (IIS Aragón), 50009, Zaragoza, Spain
| | - Mara Cruellas
- Medical Oncology Department, Vall d'Hebrón University Hospital, 08035, Barcelona, Spain
| | | | - Julián Pardo
- Health Research Institute of Aragón (IIS Aragón), 50009, Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute (ISCIII), Madrid, Spain
- Microbiology Department, Preventive Medicine and Public Health, University of Zaragoza, 50009, Zaragoza, Spain
| | - Luis Martínez-Lostao
- Microbiology Department, Preventive Medicine and Public Health, University of Zaragoza, 50009, Zaragoza, Spain
| | | | - Patricia Esteban
- Health Research Institute of Aragón (IIS Aragón), 50009, Zaragoza, Spain
| | - Irene Torres-Ramón
- Medical Oncology Department, Lozano Blesa University Hospital Clinic, 50009, Zaragoza, Spain
- Health Research Institute of Aragón (IIS Aragón), 50009, Zaragoza, Spain
| | - Alfonso Yubero
- Medical Oncology Department, Lozano Blesa University Hospital Clinic, 50009, Zaragoza, Spain
- Health Research Institute of Aragón (IIS Aragón), 50009, Zaragoza, Spain
| | - José Ramón Paño
- Health Research Institute of Aragón (IIS Aragón), 50009, Zaragoza, Spain
- ESCMID Fellow, Infectious Diseases Department, Lozano Blesa University Hospital Clinic, Zaragoza, Spain and University of Zaragoza, 50009, Zaragoza, Spain
| | - Rodrigo Lastra
- Medical Oncology Department, Lozano Blesa University Hospital Clinic, 50009, Zaragoza, Spain
- Health Research Institute of Aragón (IIS Aragón), 50009, Zaragoza, Spain
| |
Collapse
|
158
|
Neal J, Pavlakis N, Kim SW, Goto Y, Lim SM, Mountzios G, Fountzilas E, Mochalova A, Christoph DC, Bearz A, Quantin X, Palmero R, Antic V, Chun E, Edubilli TR, Lin YC, Huseni M, Ballinger M, Graupner V, Curran D, Vervaet P, Newsom-Davis T. CONTACT-01: A Randomized Phase III Trial of Atezolizumab + Cabozantinib Versus Docetaxel for Metastatic Non-Small Cell Lung Cancer After a Checkpoint Inhibitor and Chemotherapy. J Clin Oncol 2024; 42:2393-2403. [PMID: 38552197 PMCID: PMC11227305 DOI: 10.1200/jco.23.02166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/04/2023] [Accepted: 02/01/2024] [Indexed: 04/28/2024] Open
Abstract
PURPOSE Although checkpoint inhibitors have improved first-line treatment for non-small cell lung cancer (NSCLC), a therapeutic need remains for patients whose disease does not respond or who experience disease progression after anti-PD-L1/PD-1 immunotherapy. CONTACT-01 (ClinicalTrials.gov identifier: NCT04471428) evaluated atezolizumab plus cabozantinib versus docetaxel in patients with metastatic NSCLC who developed disease progression after concurrent or sequential treatment with anti-PD-L1/PD-1 and platinum-containing chemotherapy. METHODS This multicenter, open-label, phase III trial randomly assigned patients 1:1 to atezolizumab 1,200 mg intravenously once every 3 weeks (q3w) plus cabozantinib 40 mg orally once daily or docetaxel 75 mg/m2 intravenously once every 3 weeks. The primary end point was overall survival (OS). RESULTS One hundred eighty-six patients were assigned atezolizumab plus cabozantinib, and 180 docetaxel. Minimum OS follow-up was 10.9 months. Median OS was 10.7 months (95% CI, 8.8 to 12.3) with atezolizumab plus cabozantinib and 10.5 months (95% CI, 8.6 to 13.0) with docetaxel (stratified hazard ratio [HR], 0.88 [95% CI, 0.68 to 1.16]; P = .3668). Median progression-free survival was 4.6 months (95% CI, 4.1 to 5.6) and 4.0 months (95% CI, 3.1 to 4.4), respectively (stratified HR, 0.74 [95% CI, 0.59 to 0.92]). Serious adverse events (AEs) occurred in 71 (38.4%) patients receiving atezolizumab plus cabozantinib and 58 (34.7%) receiving docetaxel. Grade 3/4 treatment-related AEs occurred in 73 (39.5%) patients receiving atezolizumab plus cabozantinib and 58 (34.7%) receiving docetaxel. Grade 5 AEs occurred in 14 (7.6%) and 10 (6.0%) patients in the atezolizumab plus cabozantinib and docetaxel arms, respectively (treatment-related in four [2.2%] and one [0.6%], respectively). CONCLUSION Atezolizumab plus cabozantinib after disease progression following anti-PD-L1/PD-1 immunotherapy and platinum-containing chemotherapy for metastatic NSCLC did not improve OS compared with docetaxel. Safety was consistent with known profiles of these agents.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/mortality
- Docetaxel/therapeutic use
- Docetaxel/administration & dosage
- Docetaxel/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/mortality
- Pyridines/therapeutic use
- Pyridines/administration & dosage
- Pyridines/adverse effects
- Male
- Female
- Anilides/therapeutic use
- Anilides/administration & dosage
- Anilides/adverse effects
- Middle Aged
- Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Immune Checkpoint Inhibitors/therapeutic use
- Immune Checkpoint Inhibitors/adverse effects
- Adult
- Aged, 80 and over
- Progression-Free Survival
Collapse
Affiliation(s)
- Joel Neal
- Stanford Cancer Institute, Stanford University, Palo Alto, CA
| | - Nick Pavlakis
- Royal North Shore Hospital, University of Sydney, St Leonards, Australia
| | - Sang-We Kim
- Asan Medical Centre, University of Ulsan College of Medicine, Seoul, South Korea
| | | | - Sun Min Lim
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | - Xavier Quantin
- Montpellier Cancer Institute, Inserm U1194, University of Montpellier, Montpellier, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
159
|
Andresen NK, Røssevold AH, Borgen E, Schirmer CB, Gilje B, Garred Ø, Lømo J, Stensland M, Nordgård O, Falk RS, Mathiesen RR, Russnes HG, Kyte JA, Naume B. Circulating tumor cells in metastatic breast cancer patients treated with immune checkpoint inhibitors - a biomarker analysis of the ALICE and ICON trials. Mol Oncol 2024. [PMID: 38978352 DOI: 10.1002/1878-0261.13675] [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: 02/14/2024] [Revised: 04/16/2024] [Accepted: 05/27/2024] [Indexed: 07/10/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have been introduced in breast cancer (BC) treatment and better biomarkers are needed to predict benefit. Circulating tumor cells (CTCs) are prognostic in BC, but knowledge is limited on CTCs in the context of ICI therapy. In this study, serial sampling of CTCs (CellSearch system) was evaluated in 82 patients with metastatic BC enrolled in two randomized trials investigating ICI plus chemotherapy. Programmed death-ligand 1 (PD-L1) expression on CTCs was also measured. Patients with ≥ 2 CTCs per 7.5 mL at baseline had gene expression profiles in tumor suggestive of increased T-cell activity, including increased tumor inflammation signature (TIS) in both triple-negative (P = 0.010) and hormone receptor-positive (P = 0.024) disease. Patients with luminal A BC had higher CTC levels. The association between CTC status and outcome was most apparent 4 weeks into therapy. PD-L1 expression in CTCs was observed in 6/17 CTC-positive patients and was associated with inferior survival. In conclusion, our study indicates that CTC numbers may inform on tumor immune composition, as well as prognosis. These findings suggest a potential of using CTCs as an accessible biomarker source in BC patients treated with immunotherapy.
Collapse
Affiliation(s)
- Nikolai Kragøe Andresen
- Department of Clinical Cancer Research, Oslo University Hospital, Norway
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Norway
- Institute of Clinical Medicine, University of Oslo, Norway
| | - Andreas Hagen Røssevold
- Department of Clinical Cancer Research, Oslo University Hospital, Norway
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Norway
- Institute of Clinical Medicine, University of Oslo, Norway
| | - Elin Borgen
- Department of Pathology, Oslo University Hospital, Norway
| | | | - Bjørnar Gilje
- Department of Hematology and Oncology, Stavanger University Hospital, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, Norway
| | - Jon Lømo
- Department of Pathology, Oslo University Hospital, Norway
| | - Marius Stensland
- Department of Hematology and Oncology, Stavanger University Hospital, Norway
| | - Oddmund Nordgård
- Department of Hematology and Oncology, Stavanger University Hospital, Norway
- Department of Chemistry, Bioscience and Environmental Technology, University of Stavanger, Norway
| | - Ragnhild Sørum Falk
- Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, Norway
| | | | - Hege G Russnes
- Institute of Clinical Medicine, University of Oslo, Norway
- Department of Pathology, Oslo University Hospital, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Norway
| | - Jon Amund Kyte
- Department of Clinical Cancer Research, Oslo University Hospital, Norway
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Norway
- Faculty of Health Sciences, Oslo Metropolitan University, Norway
| | - Bjørn Naume
- Institute of Clinical Medicine, University of Oslo, Norway
- Department of Oncology, Oslo University Hospital, Norway
| |
Collapse
|
160
|
Lieber A, Makai A, Orosz Z, Kardos T, Isaac SJ, Tornyi I, Bittner N. The role of immunotherapy in early-stage and metastatic NSCLC. Pathol Oncol Res 2024; 30:1611713. [PMID: 39027681 PMCID: PMC11254634 DOI: 10.3389/pore.2024.1611713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/05/2024] [Indexed: 07/20/2024]
Abstract
In the past decade we have seen new advances and thus remarkable progress in the therapeutic options for non-small cell lung cancer (NSCLC). Among cytostatic therapies with new approaches in molecularly targeted therapies, we see new developments in a wide range of applications for immunotherapies. In this review we discuss the new potential modalities for the use of immune checkpoint inhibitors (ICIs) in the frontlines, including in early-stage (perioperative) and metastatic settings. The perioperative use of ICIs in both neoadjuvant and adjuvant settings may show benefits for patients. In early-stage NSCLC (from stage IIB and above) a multimodality approach is recommended as the gold standard for the treatment. After surgical resection platinum-based adjuvant chemotherapy has been the standard of care for many years. Based on the benefit of disease-free survival, the approval of adjuvant atezolizumab and adjuvant pembrolizumab was a significant breakthrough. In the metastatic setting, the use of immune checkpoint inhibitors with chemotherapy, regardless of PD-L1 expression or ICI alone (PD-L1 expression equal to or greater than 50%) also improves overall survival and progression-free survival.
Collapse
Affiliation(s)
- Attila Lieber
- Department of Pulmonology, University of Debrecen, Debrecen, Hungary
| | - Attila Makai
- Department of Pulmonology, University of Debrecen, Debrecen, Hungary
| | - Zsuzsanna Orosz
- Department of Pulmonology, University of Debrecen, Debrecen, Hungary
| | - Tamás Kardos
- Department of Pulmonology, University of Debrecen, Debrecen, Hungary
| | - Susil Joe Isaac
- Department of Pulmonology, University of Debrecen, Debrecen, Hungary
| | - Ilona Tornyi
- Department of Pulmonology, University of Debrecen, Debrecen, Hungary
| | - Nóra Bittner
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| |
Collapse
|
161
|
Naratornsirakul D, Chewaskulyong B, Kongkarnka S, Oranratnachai S. Comparison of treatment outcome between first-line combination immunotherapy (anti-PD-L1 or anti-PD1) with or without chemotherapy and chemotherapy alone in advanced non-small cell lung cancer patients in tertiary care hospital. Cancer Med 2024; 13:e70007. [PMID: 39030820 PMCID: PMC11257996 DOI: 10.1002/cam4.70007] [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: 05/13/2024] [Revised: 06/26/2024] [Accepted: 06/30/2024] [Indexed: 07/22/2024] Open
Abstract
BACKGROUND Despite promising outcomes of first-line immunotherapy with or without chemotherapy in advanced non-small cell lung cancer (NSCLC), limited accessibility due to reimbursement was remain the problem in low to middle income countries. This study aimed to evaluate real-world effectiveness of immunotherapy in patients with advanced NSCLC in Northern Thailand. METHOD A retrospective, single-centered cohort, was conducted. Patients with advanced NSCLC who underwent PD-L1 testing (excluding EGFR and ALK mutations) and were treated with immunotherapy or without chemotherapy or chemotherapy alone were included. The primary end point was progression-free survival (PFS). The secondary endpoints were overall survival (OS), objective response rate (ORR), and adverse events. RESULTS A total of 123 patients, of which 21 patients received immunotherapy-based regimen and 102 patients received chemotherapy alone. The median PFS was 11.9 months in immunotherapy-based group compared to 5.93 months in the chemotherapy group, with a. hazard ratio (HR) of 0.4 (95% confidence interval [CI], 0.23 to 0.68; p = 0.001). Similarly, the median OS was 26.68 months in the immunotherapy-based group and 11.21 months in the chemotherapy group, with HR of 0.42 (95% CI 0.22-0.8; p = 0.009). ORRs were significantly higher in the immunotherapy-based group, with 65% of patients showing a response compared to 32% in the chemotherapy group (p = 0.006). CONCLUSION The result of this real-world study in patients with advanced stage NSCLC indicate that first-line immunotherapy-based regimen was associated with significantly greater PFS, OS, and ORR with a safety profile consistent with pivotal studies.
Collapse
Affiliation(s)
- Danainut Naratornsirakul
- Division of Medical Oncology, Department of Internal Medicine, Faculty of MedicineChiang Mai UniversityChiang MaiThailand
| | - Busyamas Chewaskulyong
- Division of Medical Oncology, Department of Internal Medicine, Faculty of MedicineChiang Mai UniversityChiang MaiThailand
| | - Sarawut Kongkarnka
- Department of Pathology, Faculty of MedicineChiang Mai UniversityChiang MaiThailand
| | | |
Collapse
|
162
|
Papazyan T, Denis MG, Sagan C, Raimbourg J, Herbreteau G, Pons-Tostivint E. Impact of PD-L1 Expression on the Overall Survival of Caucasian Patients with Advanced EGFR-Mutant NSCLC Treated with Frontline Osimertinib. Target Oncol 2024; 19:611-621. [PMID: 38825654 DOI: 10.1007/s11523-024-01072-x] [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: 05/14/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND The treatment of advanced non-small cell lung cancer (NSCLC) harboring an oncogenic epidermal growth factor receptor mutation (EGFRm) is currently based on osimertinib, a third-generation tyrosine kinase inhibitor (TKI). High Programmed death ligand 1 (PD-L1) expression ≥ 50% demonstrated to be a negative prognostic factor, mostly among Asian populations treated with 1st/2nd generation TKI. OBJECTIVE We investigated the impact of PD-L1 expression on the progression free survival (PFS) and overall survival (OS) within a cohort of patients receiving osimertinib as first-line treatment. METHODS Our bi-centre French retrospective study included all newly diagnosed patients with an advanced EGFRm (common and uncommon) NSCLC, between May 2018 and November 2022, treated with osimertinib. The primary endpoint was OS according to tumor proportion score PD-L1 expression (low/intermediate < 50% vs high ≥ 50%). Survival analyses were performed using Kaplan-Meier method and Cox model for adjusted multivariate analysis. RESULTS Of 96 patients, median age was 71 (IQR 62-76), 70 were women (72.9%), 81 had a performance status (PS) 0-1 (84.3%). Median follow-up was 22.6 months (95% CI 20.5-24.7). Twenty patients (20.8%) had high PD-L1 expression ≥ 50%. No significant differences in baseline characteristics were observed based on PD-L1 status. Patients with PD-L1 ≥ 50% had significant shorter PFS and OS than those with PD-L1 < 50%, respectively 9.3 vs 17.5 months (p = 0.044 months) and 14.3 vs 26.0 months (p = 0.025). Multivariable adjustment for baseline characteristics found that PS ≥ 2 (HR 2.79, 95% CI 1.12-6.93, p = 0.027), PD-L1 ≥ 50% (HR 2.61, 95% CI 1.31 to 5.22, p = 0.007) and uncommon EGFR mutation (HR 4.59, 95% CI 1.95-10.80, p = <0.001) were associated with a shorter OS. Brain metastases at diagnosis and age ≥ 65 were not, respectively HR 1.66 (95% CI 0.90-3.06, p = 0.11) and HR 0.95 (95% CI 0.50-1.80, p=0.9). CONCLUSIONS Our study found that PD-L1 expression ≥ 50% was associated with a shorter OS in EGFRm NSCLC patients treated with first line osimertinib. Further research is warranted to understand the underlying molecular and cellular mechanisms of this correlation.
Collapse
Affiliation(s)
- Thomas Papazyan
- Medical oncology, Centre Hospitalier Universitaire Nantes, Nantes University, Boulevard Professeur Jacques Monod, 44800, Saint Herblain, France
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, Nantes, France
| | - Marc G Denis
- Department of Biochemistry, Centre Hospitalier Universitaire Nantes, Nantes University, 44000, Nantes, France
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, Nantes, France
| | - Christine Sagan
- Pathology Department, Centre Hospitalier Universitaire Nantes, Nantes University, 44000, Nantes, France
| | - Judith Raimbourg
- Department of Medical Oncology, Comprehensive Cancer Center, Institut de Cancérologie de L'Ouest, Saint-Herblain, France
| | - Guillaume Herbreteau
- Department of Biochemistry, Centre Hospitalier Universitaire Nantes, Nantes University, 44000, Nantes, France
| | - Elvire Pons-Tostivint
- Medical oncology, Centre Hospitalier Universitaire Nantes, Nantes University, Boulevard Professeur Jacques Monod, 44800, Saint Herblain, France.
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA, Nantes, France.
| |
Collapse
|
163
|
Ralphs E, Rault C, Calleja A, Daumont MJ, Penrod JR, Thompson M, Cheeseman S, Soares M. Algorithms to identify radiotherapy intent in unresected non-metastatic non-small-cell lung cancer: an I-O Optimise analysis. Future Oncol 2024; 20:1633-1643. [PMID: 38904271 PMCID: PMC11485750 DOI: 10.1080/14796694.2024.2363133] [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: 08/24/2023] [Accepted: 05/30/2024] [Indexed: 06/22/2024] Open
Abstract
This study aimed to develop and evaluate the performance of algorithms for identifying radiotherapy (RT) treatment intent in real-world data from patients with non-metastatic non-small-cell lung cancer (NSCLC). Using data from IPO-Porto hospital (Portugal) and the REAL-Oncology database (England), three algorithms were developed based on available RT information (#1: RT duration, #2: RT duration and type, #3: RT dose) and tested versus reference datasets. Study results showed that all three algorithms had good overall accuracy (91-100%) for patients receiving RT plus systemic anticancer therapy (SACT) and algorithms #2 and #3 also had good accuracy (>99%) for patients receiving RT alone. These algorithms could help classify treatment intent in patients with NSCLC receiving RT with or without SACT in real-world settings where intent information is missing/incomplete.
Collapse
Affiliation(s)
- Eleanor Ralphs
- Real World Solutions, IQVIA, The Point, 37 N Wharf Rd, London, W2 1AF, England
| | | | - Alan Calleja
- Real World Solutions, IQVIA, The Point, 37 N Wharf Rd, London, W2 1AF, England
| | - Melinda J Daumont
- Worldwide Health Economics & Outcomes Research, Bristol Myers Squibb, Av. de Finlande 4, 1420 Braine-L'Alleud, Belgium
| | - John R Penrod
- Worldwide Health Economics & Outcomes Research, Bristol Myers Squibb, 3551 Lawrenceville Rd, Princeton, NJ08540, USA
| | - Matthew Thompson
- REAL Oncology, Leeds Teaching Hospitals NHS Trust, Bexley Wing Cancer Centre, Beckett Street, Leeds, LS9 7TF, England
| | - Sue Cheeseman
- REAL Oncology, Leeds Teaching Hospitals NHS Trust, Bexley Wing Cancer Centre, Beckett Street, Leeds, LS9 7TF, England
| | - Marta Soares
- Department of Medical Oncology, Portuguese Oncology Institute of Porto (IPO-Porto), Rua Dr. António Bernardino e Almeida, 4200–072Porto, Portugal
| |
Collapse
|
164
|
Camerini A, Mazzoni F, Scotti V, Tibaldi C, Sbrana A, Calabrò L, Caliman E, Ciccone LP, Bernardini L, Graziani J, Grosso MA, Chella A, Allegrini G, Amoroso D, Baldini E. Efficacy and Safety of Chemotherapy after Immunotherapy in Patients with Advanced Non-Small-Cell Lung Cancer. J Clin Med 2024; 13:3642. [PMID: 38999206 PMCID: PMC11242759 DOI: 10.3390/jcm13133642] [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: 04/16/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
Background: There are currently few data about the safety and effectiveness of chemotherapy for patients with metastatic non-small-cell lung cancer (NSCLC) who have progressed from prior immunotherapy. Methods: Data from patients with consecutive stage IIIB-IV, ECOG performance status (PS) 0-2, non-small-cell lung cancer (NSCLC) treated with combination or single-agent chemotherapy following progression on an earlier immunotherapy regimen were retrospectively gathered. Recorded were baseline attributes, outcome metrics, and toxicities. The neutrophil/lymphocyte (N/L) ratio's predictive usefulness was examined through an exploratory analysis. Results: The analysis comprised one hundred subjects. The adeno/squamous carcinoma ratio was 77%/23%, the M/F ratio was 66%/34%, the ECOG PS was 0/1/≥2 47%/51%/2%, and the median PD-L1 expression was 50% (range 0-100). The median age was 67 (range 39-81) years. Prior immunotherapy included a single-agent treatment in 83% of cases, with pembrolizumab use being prevalent, and a median N/L ratio of four prior to chemotherapy. The overall median time-to-progression on previous immunotherapy was 6 months. After immunotherapy, just 33% of subjects underwent chemotherapy. A median of 4 (range 1-16) cycles of chemotherapy were administered; platinum doublets (primarily carboplatin) were delivered in only 31% of cases, vinorelbine accounted for 25%, taxanes for 25%, and gemcitabine for 8%. The median clinical benefit was 55%, while the overall response rate was 21%. The median overall survival was 5 months (range 1-22) and the median time to progression was 4 months (range 1-17). Subgroups with low and high N/L ratios were compared, but there was no discernible difference in survival. Conclusions: After immunotherapy, a small percentage of patients with advanced NSCLC had chemotherapy. Following immunotherapy advancement, chemotherapy demonstrated a moderate level of therapeutic effectiveness; no adverse concerns were noted. The effectiveness of chemotherapy following immunotherapy was not predicted by the baseline N/L ratio.
Collapse
Affiliation(s)
- Andrea Camerini
- Medical Oncology, Versilia Hospital, Azienda USL Toscana Nord-Ovest, 55041 Lido di Camaiore, Italy; (M.A.G.); (D.A.)
| | - Francesca Mazzoni
- SODc Oncologia Medica, Azienda Ospedaliero Universitaria Careggi, 50134 Firenze, Italy; (F.M.); (E.C.)
| | - Vieri Scotti
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, 50134 Firenze, Italy; (V.S.); (L.P.C.)
| | - Carmelo Tibaldi
- Medical Oncology, San Luca Hospital, Azienda USL Toscana Nord-Ovest, 55100 Lucca, Italy; (C.T.); (E.B.)
| | - Andrea Sbrana
- Pneumo-Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, 50134 Pisa, Italy; (A.S.); (A.C.)
| | - Luana Calabrò
- Medical Oncology, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
| | - Enrico Caliman
- SODc Oncologia Medica, Azienda Ospedaliero Universitaria Careggi, 50134 Firenze, Italy; (F.M.); (E.C.)
| | - Lucia Pia Ciccone
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, 50134 Firenze, Italy; (V.S.); (L.P.C.)
| | - Laura Bernardini
- UO Oncologia Medica 2 Universitaria, Ospedale S. Chiara, Azienda Ospedaliero-Universitaria Pisana, 50134 Pisa, Italy; (L.B.); (J.G.)
| | - Jessica Graziani
- UO Oncologia Medica 2 Universitaria, Ospedale S. Chiara, Azienda Ospedaliero-Universitaria Pisana, 50134 Pisa, Italy; (L.B.); (J.G.)
| | - Maria Antonietta Grosso
- Medical Oncology, Versilia Hospital, Azienda USL Toscana Nord-Ovest, 55041 Lido di Camaiore, Italy; (M.A.G.); (D.A.)
| | - Antonio Chella
- Pneumo-Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, 50134 Pisa, Italy; (A.S.); (A.C.)
| | - Giacomo Allegrini
- Medical Oncology, Spedali Riuniti Livorno, Azienda USL Toscana Nord-Ovest, 57124 Livorno, Italy;
| | - Domenico Amoroso
- Medical Oncology, Versilia Hospital, Azienda USL Toscana Nord-Ovest, 55041 Lido di Camaiore, Italy; (M.A.G.); (D.A.)
| | - Editta Baldini
- Medical Oncology, San Luca Hospital, Azienda USL Toscana Nord-Ovest, 55100 Lucca, Italy; (C.T.); (E.B.)
| |
Collapse
|
165
|
Ottaiano A, Grassi F, Sirica R, Genito E, Ciani G, Patanè V, Monti R, Belfiore MP, Urraro F, Santorsola M, Ponsiglione AM, Montella M, Cappabianca S, Reginelli A, Sansone M, Savarese G, Grassi R. Associations between Radiomics and Genomics in Non-Small Cell Lung Cancer Utilizing Computed Tomography and Next-Generation Sequencing: An Exploratory Study. Genes (Basel) 2024; 15:803. [PMID: 38927739 PMCID: PMC11202615 DOI: 10.3390/genes15060803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Radiomics, an evolving paradigm in medical imaging, involves the quantitative analysis of tumor features and demonstrates promise in predicting treatment responses and outcomes. This study aims to investigate the predictive capacity of radiomics for genetic alterations in non-small cell lung cancer (NSCLC). METHODS This exploratory, observational study integrated radiomic perspectives using computed tomography (CT) and genomic perspectives through next-generation sequencing (NGS) applied to liquid biopsies. Associations between radiomic features and genetic mutations were established using the Area Under the Receiver Operating Characteristic curve (AUC-ROC). Machine learning techniques, including Support Vector Machine (SVM) classification, aim to predict genetic mutations based on radiomic features. The prognostic impact of selected gene variants was assessed using Kaplan-Meier curves and Log-rank tests. RESULTS Sixty-six patients underwent screening, with fifty-seven being comprehensively characterized radiomically and genomically. Predominantly males (68.4%), adenocarcinoma was the prevalent histological type (73.7%). Disease staging is distributed across I/II (38.6%), III (31.6%), and IV (29.8%). Significant correlations were identified with mutations of ROS1 p.Thr145Pro (shape_Sphericity), ROS1 p.Arg167Gln (glszm_ZoneEntropy, firstorder_TotalEnergy), ROS1 p.Asp2213Asn (glszm_GrayLevelVariance, firstorder_RootMeanSquared), and ALK p.Asp1529Glu (glcm_Imc1). Patients with the ROS1 p.Thr145Pro variant demonstrated markedly shorter median survival compared to the wild-type group (9.7 months vs. not reached, p = 0.0143; HR: 5.35; 95% CI: 1.39-20.48). CONCLUSIONS The exploration of the intersection between radiomics and cancer genetics in NSCLC is not only feasible but also holds the potential to improve genetic predictions and enhance prognostic accuracy.
Collapse
Affiliation(s)
- Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, 80131 Naples, Italy; (A.O.); (M.S.)
| | - Francesca Grassi
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (E.G.); (G.C.); (V.P.); (R.M.); (M.P.B.); (F.U.); (S.C.); (A.R.); (R.G.)
| | - Roberto Sirica
- AMES—Centro Polidiagnostico Strumentale, SRL, 80013 Naples, Italy; (R.S.); (G.S.)
| | - Emanuela Genito
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (E.G.); (G.C.); (V.P.); (R.M.); (M.P.B.); (F.U.); (S.C.); (A.R.); (R.G.)
| | - Giovanni Ciani
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (E.G.); (G.C.); (V.P.); (R.M.); (M.P.B.); (F.U.); (S.C.); (A.R.); (R.G.)
| | - Vittorio Patanè
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (E.G.); (G.C.); (V.P.); (R.M.); (M.P.B.); (F.U.); (S.C.); (A.R.); (R.G.)
| | - Riccardo Monti
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (E.G.); (G.C.); (V.P.); (R.M.); (M.P.B.); (F.U.); (S.C.); (A.R.); (R.G.)
| | - Maria Paola Belfiore
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (E.G.); (G.C.); (V.P.); (R.M.); (M.P.B.); (F.U.); (S.C.); (A.R.); (R.G.)
| | - Fabrizio Urraro
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (E.G.); (G.C.); (V.P.); (R.M.); (M.P.B.); (F.U.); (S.C.); (A.R.); (R.G.)
| | - Mariachiara Santorsola
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, 80131 Naples, Italy; (A.O.); (M.S.)
| | - Alfonso Maria Ponsiglione
- Department of Electrical Engineering and Information Technology, University of Naples “Federico II”, 80125 Naples, Italy; (A.M.P.); (M.S.)
| | - Marco Montella
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Salvatore Cappabianca
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (E.G.); (G.C.); (V.P.); (R.M.); (M.P.B.); (F.U.); (S.C.); (A.R.); (R.G.)
| | - Alfonso Reginelli
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (E.G.); (G.C.); (V.P.); (R.M.); (M.P.B.); (F.U.); (S.C.); (A.R.); (R.G.)
| | - Mario Sansone
- Department of Electrical Engineering and Information Technology, University of Naples “Federico II”, 80125 Naples, Italy; (A.M.P.); (M.S.)
| | - Giovanni Savarese
- AMES—Centro Polidiagnostico Strumentale, SRL, 80013 Naples, Italy; (R.S.); (G.S.)
| | - Roberta Grassi
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (E.G.); (G.C.); (V.P.); (R.M.); (M.P.B.); (F.U.); (S.C.); (A.R.); (R.G.)
| |
Collapse
|
166
|
Tóth LJ, Mokánszki A, Méhes G. The rapidly changing field of predictive biomarkers of non-small cell lung cancer. Pathol Oncol Res 2024; 30:1611733. [PMID: 38953007 PMCID: PMC11215025 DOI: 10.3389/pore.2024.1611733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 06/04/2024] [Indexed: 07/03/2024]
Abstract
Lung cancer is a leading cause of cancer-related death worldwide in both men and women, however mortality in the US and EU are recently declining in parallel with the gradual cut of smoking prevalence. Consequently, the relative frequency of adenocarcinoma increased while that of squamous and small cell carcinomas declined. During the last two decades a plethora of targeted drug therapies have appeared for the treatment of metastasizing non-small cell lung carcinomas (NSCLC). Personalized oncology aims to precisely match patients to treatments with the highest potential of success. Extensive research is done to introduce biomarkers which can predict the effectiveness of a specific targeted therapeutic approach. The EGFR signaling pathway includes several sufficient targets for the treatment of human cancers including NSCLC. Lung adenocarcinoma may harbor both activating and resistance mutations of the EGFR gene, and further, mutations of KRAS and BRAF oncogenes. Less frequent but targetable genetic alterations include ALK, ROS1, RET gene rearrangements, and various alterations of MET proto-oncogene. In addition, the importance of anti-tumor immunity and of tumor microenvironment has become evident recently. Accumulation of mutations generally trigger tumor specific immune defense, but immune protection may be upregulated as an aggressive feature. The blockade of immune checkpoints results in potential reactivation of tumor cell killing and induces significant tumor regression in various tumor types, such as lung carcinoma. Therapeutic responses to anti PD1-PD-L1 treatment may correlate with the expression of PD-L1 by tumor cells. Due to the wide range of diagnostic and predictive features in lung cancer a plenty of tests are required from a single small biopsy or cytology specimen, which is challenged by major issues of sample quantity and quality. Thus, the efficacy of biomarker testing should be warranted by standardized policy and optimal material usage. In this review we aim to discuss major targeted therapy-related biomarkers in NSCLC and testing possibilities comprehensively.
Collapse
Affiliation(s)
- László József Tóth
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | | |
Collapse
|
167
|
Fujiwara Y, Shimomura K, Yamaguchi T, Shimizu J, Watanabe N, Matsuzawa R, Murotani K, Horio Y. The incidence of drug-induced interstitial lung disease caused by epidermal growth factor receptor tyrosine kinase inhibitors or immune checkpoint inhibitors in patients with non-small cell lung cancer in presence and absence of vascular endothelial growth factor inhibitors: a systematic review. Front Oncol 2024; 14:1419256. [PMID: 38919534 PMCID: PMC11196607 DOI: 10.3389/fonc.2024.1419256] [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: 04/17/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
Interstitial lung disease (ILD) or pneumonitis caused by epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKI) or immune checkpoint inhibitors (ICI) is a major concern in the treatment of non-small cell lung cancer (NSCLC). Whether the addition of vascular endothelial growth factor (VEGF) and VEGF receptor (VEGFR) inhibitors can reduce the incidence of drug-induced ILD remains unclear. We conducted a systematic review to assess the incidence of ILD induced by EGFR-TKIs or ICIs in the presence or absence of VEGF/VEGFR inhibitors in relevant randomized trials between January 2009 and October 2023. The primary outcome was the odds ratio for the incidence of ILD in all patients worldwide and Asians. Secondary outcomes were the odds ratios (ORs) of the incidence at grade-3 or higher ILD in all patients worldwide and Asians. We identified 13 randomized studies, one sub-analysis in the EGFR-TKI group, and three randomized studies in the ICI group. In the EGFR-TKI group, the OR of ILD incidence at any grade with VEGF/VEGFR inhibitors was 0.54 (95% CI, 0.32-0.90; p = 0.02), which represented a significantly lower incidence than that without VEGF/VEGFR inhibitors. Contrarily, the OR of ILD incidence at grade ≥ 3 with VEGF/VEGFR inhibitors was 1.00 (95% CI, 0.43-2.36; p = 0.99). In all subjects in the ICI group, the OR of ILD incidence at any grade with VEGF/VEGFR inhibitors was 0.78 (95% CI, 0.51-1.21; p = 0.27). The systematic review demonstrated that the addition of VEGF/VEGFR inhibitors could reduce the incidence of drug-induced ILD at any grade caused by EGFR-TKI in patients with NSCLC but could not reduce that at grade ≥ 3. The ILD induced by ICIs remains undetermined owing to the limited number of randomized trials for which ILD data are available. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=409534, identifier CRD42023409534.
Collapse
Affiliation(s)
- Yutaka Fujiwara
- Department of Thoracic Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Kazuhiro Shimomura
- Department of Pharmacy, Aichi Cancer Center, Nagoya, Japan
- Biostatistics Center, Kurume University, Kurume, Japan
| | - Teppei Yamaguchi
- Department of Thoracic Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Junichi Shimizu
- Department of Thoracic Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Naohiro Watanabe
- Department of Thoracic Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Reiko Matsuzawa
- Department of Thoracic Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Kenta Murotani
- Biostatistics Center, Kurume University, Kurume, Japan
- School of Medical Technology, Kurume University, Kurume, Japan
| | - Yoshitsugu Horio
- Department of Thoracic Oncology, Aichi Cancer Center, Nagoya, Japan
| |
Collapse
|
168
|
Li X, Feng X, Zhou J, Luo Y, Chen X, Zhao J, Chen H, Xiong G, Luo G. A muti-modal feature fusion method based on deep learning for predicting immunotherapy response. J Theor Biol 2024; 586:111816. [PMID: 38589007 DOI: 10.1016/j.jtbi.2024.111816] [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: 10/21/2023] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Immune checkpoint therapy (ICT) has greatly improved the survival of cancer patients in the past few years, but only a small number of patients respond to ICT. To predict ICT response, we developed a multi-modal feature fusion model based on deep learning (MFMDL). This model utilizes graph neural networks to map gene-gene relationships in gene networks to low dimensional vector spaces, and then fuses biological pathway features and immune cell infiltration features to make robust predictions of ICT. We used five datasets to validate the predictive performance of the MFMDL. These five datasets span multiple types of cancer, including melanoma, lung cancer, and gastric cancer. We found that the prediction performance of multi-modal feature fusion model based on deep learning is superior to other traditional ICT biomarkers, such as ICT targets or tumor microenvironment-associated markers. In addition, we also conducted ablation experiments to demonstrate the necessity of fusing different modal features, which can improve the prediction accuracy of the model.
Collapse
Affiliation(s)
- Xiong Li
- School of Software, East China Jiaotong University, Nanchang 330013, China
| | - Xuan Feng
- School of Software, East China Jiaotong University, Nanchang 330013, China
| | - Juan Zhou
- School of Software, East China Jiaotong University, Nanchang 330013, China
| | - Yuchao Luo
- School of Software, East China Jiaotong University, Nanchang 330013, China
| | - Xiao Chen
- School of Software, East China Jiaotong University, Nanchang 330013, China
| | - Jiapeng Zhao
- School of Software, East China Jiaotong University, Nanchang 330013, China
| | - Haowen Chen
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, China.
| | - Guoming Xiong
- School of Software, East China Jiaotong University, Nanchang 330013, China
| | - Guoliang Luo
- School of Software, East China Jiaotong University, Nanchang 330013, China
| |
Collapse
|
169
|
Cavdar E, Karaboyun K, Kara K. Comprehensive analysis of the prognostic role of laboratory indices in advanced lung cancer patients. Asia Pac J Clin Oncol 2024. [PMID: 38837742 DOI: 10.1111/ajco.14092] [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: 01/25/2024] [Revised: 05/16/2024] [Accepted: 05/26/2024] [Indexed: 06/07/2024]
Abstract
OBJECTIVE Lung cancer, the most common cause of cancer-related death, is diagnosed mostly in advanced stages, and 5-year survival is approximately 5.8%. It is critical to identify reliable prognostic factors to optimize treatment responses, guide therapeutic strategies and pave the way to new research. In this study, we aimed to investigate the strongest prognostic factors for advanced non-small cell lung cancer (NSCLC). METHODS We retrospectively analyzed 278 patients with NSCLC. We evaluated the association between potential prognostic factors and overall survival (OS) times using Kaplan-Meier analysis and Cox regression analysis. RESULTS The median OS in all patients was 15.3 months. In univariate analysis, gender, histologic type, performance status, immunotherapy, radiotherapy, hemoglobin level, serum albumin, sodium-globulin ratio (SGR), neutrophil-lymphocyte ratio (NLR), systemic immune inflammation index (SII), hemoglobin-albumin-lymphocyte-platelet score (HALP), and advanced lung cancer index (ALI) were associated with survival. Models were established for multivariate analyses. In the models, NLR, SGR, HALP, immunotherapy, radiotherapy, and Eastern Cooperative Oncology Group (ECOG) performance status showed independent prognostic features (p < 0.001, p = 0.003, p = 0.002, p < 0.001, p = 0.010, and p = 0.025, respectively). In addition, in the subgroup analysis, prognostic indexes (NLR, SGR, and HALP) were found to have a prognostic effect on survival in multiple subgroups. CONCLUSIONS Pretreatment NLR, SGR, HALP, immunotherapy, radiotherapy, and ECOG performance status are independent prognostic factors for advanced NSCLC patients. These prognostic factors can be used in clinical practice as easily accessible, simple, and useful tools for clinicians.
Collapse
Affiliation(s)
- Eyyüp Cavdar
- Department of Medical Oncology, Training and Research Hospital, Adiyaman University, Adiyaman, Turkey
| | - Kubilay Karaboyun
- Department of Medical Oncology, Training and Research Hospital, Agri Ibrahim Cecen University, Agri, Turkey
| | - Kaan Kara
- Department of Chest Disease, Yedikule Chest Diseases and Thoracic Surgery Training and Research Hospital, University of Health Sciences, İstanbul, Turkey
| |
Collapse
|
170
|
Marc Malovrh M, Adamic K. Unravelling the lung cancer diagnostic pathway: identifying gaps and opportunities for improvement. Radiol Oncol 2024; 58:268-278. [PMID: 38613841 PMCID: PMC11165972 DOI: 10.2478/raon-2024-0025] [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: 12/18/2023] [Accepted: 01/20/2024] [Indexed: 04/15/2024] Open
Abstract
BACKGROUND A fast and well-organized complex diagnostic process is important for better success in the treatment of lung cancer patients. The aim of our study was to reveal the gaps and inefficiencies in the diagnostic process and to suggest improvement strategies in a single tertiary centre in Slovenia. PATIENTS AND METHODS We employed a comprehensive approach to carefully dissect all the steps in the diagnostic journey for individuals suspected of having lung cancer. We gathered and analysed information from employees and patients involved in the process by dedicated questionnaires. Further, we analysed the patients' data and calculated the diagnostic intervals for patients in two different periods. RESULTS The major concerns among employees were stress and excessive administrative work. The important result of the visual journey and staff reports was the design of electronic diagnostic clinical pathway (eDCP), which could substantially increase safety and efficacy by diminishing the administrative burden of the employees. The patients were generally highly satisfied with diagnostic journey, but reported too long waiting times. By analysing two time periods, we revealed that diagnostic intervals exceeded the recommended timelines and got importantly shorter after two interventions - strengthening the diagnostic team and specially by purchase of additional PET-CT machine (the average time from general practitioner (GP) referral to the multidisciplinary treatment board (MDTB) decision was 50.8 [± 3.0] prior and 37.1 [± 2.3] days after the interventions). CONCLUSIONS The study illuminated opportunities for refining the diagnostic journey for lung cancer patients, underscoring the importance of both administrative and capacity-related enhancements.
Collapse
Affiliation(s)
- Mateja Marc Malovrh
- University Clinic for Respiratory and Allergic Diseases Golnik, Golnik, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Katja Adamic
- University Clinic for Respiratory and Allergic Diseases Golnik, Golnik, Slovenia
| |
Collapse
|
171
|
Frost N, Reck M. Non-Small Cell Lung Cancer Metastatic Without Oncogenic Alterations. Am Soc Clin Oncol Educ Book 2024; 44:e432524. [PMID: 38669613 DOI: 10.1200/edbk_432524] [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: 04/28/2024]
Abstract
This overview provides a thorough review of current treatment approaches for first-line management of nononcogenic addicted non-small cell lung cancer. We also address pertinent clinical decision-making queries encountered in everyday practice, such as the optimal treatment strategy for PD-L1-high patients, predictive factors for response to immune checkpoint inhibitors (ICI) both in terms of patient and cancer characteristics, the potential benefits of dual checkpoint blockade, and the unresolved issue of safe discontinuation strategies for long-term responders. Around one in five patients falls into this latter category while the majority develop either primary or acquired resistance to ICI-based first-line therapy, necessitating effective subsequent lines of treatment. Docetaxel, with or without combination of antiangiogenic agents, serves as the backbone of treatment, although evidence in the post-ICI setting is limited. Given that an inflamed tumor microenvironment (TME) is crucial for ICI responses, targeting the TME in cases of acquired resistance alongside continued ICI administration appears rational, although clinical trials so far have failed to confirm this hypothesis. Antibody-drug conjugates have emerged as a promising treatment modality, offering the potential for reduced toxicity and improved efficacy by targeting specific cancer antigens. Moreover, several chemotherapy-free approaches are currently under investigation for treatment-naïve patients, including alternative ICI and drugs targeting epitopes on both cancer and immune cells.
Collapse
Affiliation(s)
- Nikolaj Frost
- Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Infectious Diseases and Pulmonary Medicine, Berlin, Germany
| | - Martin Reck
- Department of Thoracic Oncology, Airway Research Center North, German Center for Lung Research, LungenClinic, Grosshansdorf, Germany
| |
Collapse
|
172
|
Scardaci R, Berlinska E, Scaparone P, Vietti Michelina S, Garbo E, Novello S, Santamaria D, Ambrogio C. Novel RAF-directed approaches to overcome current clinical limits and block the RAS/RAF node. Mol Oncol 2024; 18:1355-1377. [PMID: 38362705 PMCID: PMC11161739 DOI: 10.1002/1878-0261.13605] [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: 07/24/2023] [Revised: 11/30/2023] [Accepted: 01/30/2024] [Indexed: 02/17/2024] Open
Abstract
Mutations in the RAS-RAF-MEK-ERK pathway are frequent alterations in cancer and RASopathies, and while RAS oncogene activation alone affects 19% of all patients and accounts for approximately 3.4 million new cases every year, less frequent alterations in the cascade's downstream effectors are also involved in cancer etiology. RAS proteins initiate the signaling cascade by promoting the dimerization of RAF kinases, which can act as oncoproteins as well: BRAFV600E is the most common oncogenic driver, mutated in the 8% of all malignancies. Research in this field led to the development of drugs that target the BRAFV600-like mutations (Class I), which are now utilized in clinics, but cause paradoxical activation of the pathway and resistance development. Furthermore, they are ineffective against non-BRAFV600E malignancies that dimerize and could be either RTK/RAS independent or dependent (Class II and III, respectively), which are still lacking an effective treatment. This review discusses the recent advances in anti-RAF therapies, including paradox breakers, dimer-inhibitors, immunotherapies, and other novel approaches, critically evaluating their efficacy in overcoming the therapeutic limitations, and their putative role in blocking the RAS pathway.
Collapse
Affiliation(s)
- Rossella Scardaci
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Ewa Berlinska
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Pietro Scaparone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Sandra Vietti Michelina
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Edoardo Garbo
- Department of OncologyUniversity of Torino, San Luigi HospitalOrbassanoItaly
| | - Silvia Novello
- Department of OncologyUniversity of Torino, San Luigi HospitalOrbassanoItaly
| | - David Santamaria
- Centro de Investigación del CáncerCSIC‐Universidad de SalamancaSpain
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| |
Collapse
|
173
|
Yang JCH, Han B, De La Mora Jiménez E, Lee JS, Koralewski P, Karadurmus N, Sugawara S, Livi L, Basappa NS, Quantin X, Dudnik J, Ortiz DM, Mekhail T, Okpara CE, Dutcus C, Zimmer Z, Samkari A, Bhagwati N, Csőszi T. Pembrolizumab With or Without Lenvatinib for First-Line Metastatic NSCLC With Programmed Cell Death-Ligand 1 Tumor Proportion Score of at least 1% (LEAP-007): A Randomized, Double-Blind, Phase 3 Trial. J Thorac Oncol 2024; 19:941-953. [PMID: 38159809 DOI: 10.1016/j.jtho.2023.12.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/22/2023] [Accepted: 12/25/2023] [Indexed: 01/03/2024]
Abstract
INTRODUCTION Lenvatinib plus pembrolizumab was found to have antitumor activity and acceptable safety in previously treated metastatic NSCLC. We evaluated first-line lenvatinib plus pembrolizumab versus placebo plus pembrolizumab in metastatic NSCLC in the LEAP-007 study (NCT03829332/NCT04676412). METHODS Patients with previously untreated stage IV NSCLC with programmed cell death-ligand 1 tumor proportion score of at least 1% without targetable EGFR/ROS1/ALK aberrations were randomized 1:1 to lenvatinib 20 mg or placebo once daily; all patients received pembrolizumab 200 mg every 3 weeks for up to 35 cycles. Primary end points were progression-free survival (PFS) per Response Evaluation Criteria in Solid Tumors version 1.1 and overall survival (OS). We report results from a prespecified nonbinding futility analysis of OS performed at the fourth independent data and safety monitoring committee review (futility bound: one-sided p < 0.4960). RESULTS A total of 623 patients were randomized. At median follow-up of 15.9 months, median (95% confidence interval [CI]) OS was 14.1 (11.4‒19.0) months in the lenvatinib plus pembrolizumab group versus 16.4 (12.6‒20.6) months in the placebo plus pembrolizumab group (hazard ratio = 1.10 [95% CI: 0.87‒1.39], p = 0.79744 [futility criterion met]). Median (95% CI) PFS was 6.6 (6.1‒8.2) months versus 4.2 (4.1‒6.2) months, respectively (hazard ratio = 0.78 [95% CI: 0.64‒0.95]). Grade 3 to 5 treatment-related adverse events occurred in 57.9% of patients (179 of 309) versus 24.4% (76 of 312). Per data and safety monitoring committee recommendation, the study was unblinded and lenvatinib and placebo were discontinued. CONCLUSIONS Lenvatinib plus pembrolizumab did not have a favorable benefit‒risk profile versus placebo plus pembrolizumab. Pembrolizumab monotherapy remains an approved treatment option in many regions for first-line metastatic NSCLC with programmed cell death-ligand 1 tumor proportion score of at least 1% without EGFR/ALK alterations.
Collapse
Affiliation(s)
- James Chih-Hsin Yang
- Department of Oncology, National Taiwan University Hospital and National Taiwan University Cancer Center, Taipei, Taiwan, Republic of China.
| | - Baohui Han
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | | | - Jong-Seok Lee
- Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | | | | | - Shunichi Sugawara
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Miyagi, Japan
| | - Lorenzo Livi
- Department of Experimental and Biomedical Sciences Mario Serio, University of Florence and Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Naveen S Basappa
- Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | - Xavier Quantin
- IRCM, INSERM, University of Montpellier, ICM, Montpellier, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Li Y, Sun H, Bai C, Hu Y, Tang J, Zhang Y, Chen J, Zhong Z, He Y, Hu K, Yang J. Dihydroartemisinin inhibits tumor progress via blocking ROR1-induced STAT3-activation in non-small cell lung cancer. Int Immunopharmacol 2024; 133:112157. [PMID: 38678671 DOI: 10.1016/j.intimp.2024.112157] [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: 01/06/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
In non-small cell lung cancer (NSCLC), identifying a component with certain molecular targets can aid research on cancer treatment. Dihydroartemisinin (DHA) is a semisynthetic derivative of artemisinin which induced the anti-cancer effects via the STAT3 signaling pathway, but the underlying molecular mechanism is still elusive. In this study, we first proved that DHA prohibits the growth of tumors both in vitro and in vivo. Data from transcriptomics showed that DHA reduced the expression level of the genes involved in cell cycle-promoting and anti-apoptosis, and most importantly, DHA restricted the expression level of receptor tyrosine kinase-like orphan receptor 1 (ROR1) which has been reported to have abnormal expression on tumor cells and had close interaction with STAT3 signaling. Then, we performed comprehensive experiments and found that DHA remarkably decreased the expression of ROR1 at both mRNA and protein levels and it also diminished the phosphorylation level of STAT3 in NSCLC cell lines. In addition, our data showed that exogenously introduced ROR1 could significantly enhance the phosphorylation of STAT3 while blocking ROR1 had the opposite effects indicating that ROR1 plays a critical role in promoting the activity of STAT3 signaling. Finally, we found that ROR1 overexpression could partially reverse the decreased activity of STAT3 induced by DHA which indicates that DHA-induced anti-growth signaling is conferred, at least in part, through blocking ROR1-mediated STAT3 activation. In summary, our study indicates that in NSCLC, ROR1 could be one of the critical molecular targets mediating DHA-induced STAT3 retardation.
Collapse
Affiliation(s)
- Yanping Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - Haoyi Sun
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - Caihong Bai
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - Youfan Hu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - Jingyi Tang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - Yuxi Zhang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - Jilan Chen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - Zhanqiong Zhong
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - Yuping He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - Kaifeng Hu
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| | - Jiahui Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| |
Collapse
|
175
|
Karlsen W, Akily L, Mierzejewska M, Teodorczyk J, Bandura A, Zaucha R, Cytawa W. Is 18F-FDG-PET/CT an Optimal Imaging Modality for Detecting Immune-Related Adverse Events after Immune-Checkpoint Inhibitor Therapy? Pros and Cons. Cancers (Basel) 2024; 16:1990. [PMID: 38893111 PMCID: PMC11171385 DOI: 10.3390/cancers16111990] [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: 04/14/2024] [Revised: 05/15/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
Immunotherapy with immune checkpoint inhibitors (ICIs) has revolutionized contemporary oncology, presenting efficacy in various solid tumors and lymphomas. However, ICIs may potentially overstimulate the immune system, leading to immune-related adverse events (irAEs). IrAEs may affect multiple organs, such as the colon, stomach, small intestine, kidneys, skin, lungs, joints, liver, lymph nodes, bone marrow, brain, heart, and endocrine glands (e.g., pancreas, thyroid, or adrenal glands), exhibiting autoimmune inflammation. 18F-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) is commonly used in oncology for staging and assessment of therapy responses, but it may also serve as a tool for detecting irAEs. This review aims to present various patterns of metabolic activation associated with irAEs due to ICI treatment, identifiable through 18F-FDG PET/CT. It describes the advantages of early detection of irAEs, but also presents the challenges in differentiating them from tumor progression. It also delves into aspects of molecular response assessment within the context of pseudoprogression and hyperprogression, along with typical imaging findings related to these phenomena. Lastly, it summarizes the role of functional PET imaging in oncological immunotherapy, speculating on its future significance and limitations.
Collapse
Affiliation(s)
- William Karlsen
- Students’ Scientific Circle Department of Nuclear Medicine, Medical University of Gdańsk, 80-952 Gdańsk, Poland; (W.K.); (L.A.)
| | - Lin Akily
- Students’ Scientific Circle Department of Nuclear Medicine, Medical University of Gdańsk, 80-952 Gdańsk, Poland; (W.K.); (L.A.)
| | - Monika Mierzejewska
- Department of Nuclear Medicine, Medical University of Gdańsk, 80-952 Gdańsk, Poland; (M.M.); (J.T.)
| | - Jacek Teodorczyk
- Department of Nuclear Medicine, Medical University of Gdańsk, 80-952 Gdańsk, Poland; (M.M.); (J.T.)
| | - Artur Bandura
- Department of Clinical Oncology and Radiotherapy, Medical University of Gdańsk, 80-952 Gdańsk, Poland; (A.B.); (R.Z.)
| | - Renata Zaucha
- Department of Clinical Oncology and Radiotherapy, Medical University of Gdańsk, 80-952 Gdańsk, Poland; (A.B.); (R.Z.)
| | - Wojciech Cytawa
- Department of Nuclear Medicine, Medical University of Gdańsk, 80-952 Gdańsk, Poland; (M.M.); (J.T.)
| |
Collapse
|
176
|
Di Pressa F, Perrone F, Benini A, Lohr F, Tiseo M, Bruni A. Management of oligometastatic and oligoprogressive epidermal growth factor receptor mutated non-small cell lung cancer patients: state of the art of a combined approach. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:449-464. [PMID: 38966183 PMCID: PMC11220311 DOI: 10.37349/etat.2024.00228] [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: 10/06/2023] [Accepted: 02/04/2024] [Indexed: 07/06/2024] Open
Abstract
Recently, the development of targeted therapy approaches such as those based on tyrosine kinase inhibitor (TKI) greatly improved the clinical outcomes of patients affected by oncogene addicted advanced non-small cell lung cancer (NSCLC). Similarly, the improvement of radiation therapy techniques has permitted to deliver high radiation doses to a limited number of metastatic target lesions (oligopersistent or oligoprogressive), with limited high-dose normal tissue exposure that leads to low severe toxicity rates. The aim of this narrative review was to provide an overview of the currently established definition of oligometastatic and oligoprogressive disease, to define first line and subsequent lines targeted therapies and the role of consolidative non-invasive local ablative treatments (LATs) in these settings. The potential benefit of local treatment (LT) such as radiotherapy (RT) or surgery might be represented by an overall reduction of switching to subsequent systemic treatments lowering the risk of further systemic dissemination. Further randomized clinical trials will clarify the role of LT and their correct timing in relation to systemic targeted therapies.
Collapse
Affiliation(s)
- Francesca Di Pressa
- Radiation Therapy Unit, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Fabiana Perrone
- Medical Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Anna Benini
- Radiation Therapy Unit, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| | - Frank Lohr
- Proton Therapy Unit, APSS Trento and CISMed, University of Trento, 38100 Trento, Italy
| | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
- Department of Medicine and Surgery, University Hospital of Parma, 43126 Parma, Italy
| | - Alessio Bruni
- Radiation Therapy Unit, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy
| |
Collapse
|
177
|
Xiao B, Li G, Gulizeba H, Liu H, Sima X, Zhou T, Huang Y. Choline metabolism reprogramming mediates an immunosuppressive microenvironment in non-small cell lung cancer (NSCLC) by promoting tumor-associated macrophage functional polarization and endothelial cell proliferation. J Transl Med 2024; 22:442. [PMID: 38730286 PMCID: PMC11084143 DOI: 10.1186/s12967-024-05242-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/27/2024] [Indexed: 05/12/2024] Open
Abstract
INTRODUCTION Lung cancer is a prevalent malignancy globally, and immunotherapy has revolutionized its treatment. However, resistance to immunotherapy remains a challenge. Abnormal cholinesterase (ChE) activity and choline metabolism are associated with tumor oncogenesis, progression, and poor prognosis in multiple cancers. Yet, the precise mechanism underlying the relationship between ChE, choline metabolism and tumor immune microenvironment in lung cancer, and the response and resistance of immunotherapy still unclear. METHODS Firstly, 277 advanced non-small cell lung cancer (NSCLC) patients receiving first-line immunotherapy in Sun Yat-sen University Cancer Center were enrolled in the study. Pretreatment and the alteration of ChE after 2 courses of immunotherapy and survival outcomes were collected. Kaplan-Meier survival and cox regression analysis were performed, and nomogram was conducted to identify the prognostic and predicted values. Secondly, choline metabolism-related genes were screened using Cox regression, and a prognostic model was constructed. Functional enrichment analysis and immune microenvironment analysis were also conducted. Lastly, to gain further insights into potential mechanisms, single-cell analysis was performed. RESULTS Firstly, baseline high level ChE and the elevation of ChE after immunotherapy were significantly associated with better survival outcomes for advanced NSCLC. Constructed nomogram based on the significant variables from the multivariate Cox analysis performed well in discrimination and calibration. Secondly, 4 choline metabolism-related genes (MTHFD1, PDGFB, PIK3R3, CHKB) were screened and developed a risk signature that was found to be related to a poorer prognosis. Further analysis revealed that the choline metabolism-related genes signature was associated with immunosuppressive tumor microenvironment, immune escape and metabolic reprogramming. scRNA-seq showed that MTHFD1 was specifically distributed in tumor-associated macrophages (TAMs), mediating the differentiation and immunosuppressive functions of macrophages, which may potentially impact endothelial cell proliferation and tumor angiogenesis. CONCLUSION Our study highlights the discovery of ChE as a prognostic marker in advanced NSCLC, suggesting its potential for identifying patients who may benefit from immunotherapy. Additionally, we developed a prognostic signature based on choline metabolism-related genes, revealing the correlation with the immunosuppressive microenvironment and uncovering the role of MTHFD1 in macrophage differentiation and endothelial cell proliferation, providing insights into the intricate workings of choline metabolism in NSCLC pathogenesis.
Collapse
Affiliation(s)
- Bijing Xiao
- Medical Oncology Department, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Guanjun Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, No. 1023-1063, Shatai Southern Road, Baiyun District, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Haimiti Gulizeba
- Medical Oncology Department, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Hong Liu
- Medical Oncology Department, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xiaoxian Sima
- Medical Oncology Department, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Ting Zhou
- Medical Oncology Department, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China.
| | - Yan Huang
- Medical Oncology Department, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
178
|
Rodriguez-Abreu D, Bosch-Barrera J, Gray JE, Ahn MJ, Johnson M, Yu X, Mohammad S, Chen X, Todd T, Kim J, Reck M. STAR-121: A Phase III Randomized Study of Domvanalimab and Zimberelimab in Combination With Chemotherapy Versus Pembrolizumab With Chemotherapy in Untreated Metastatic Non-Small Cell Lung Cancer With No Actionable Gene Alterations. Clin Lung Cancer 2024; 25:274-279. [PMID: 38310035 DOI: 10.1016/j.cllc.2023.12.010] [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: 12/13/2023] [Accepted: 12/17/2023] [Indexed: 02/05/2024]
Abstract
INTRODUCTION Dual inhibition with a T-cell immunoreceptor with immunoglobulin and ITIM domains plus programmed death (ligand)-1 (PD[L]-1) inhibitors, with or without chemotherapy, is an emerging therapeutic strategy in metastatic non-small cell lung cancer (mNSCLC). The STAR-121 (NCT05502237) phase III, global, randomized, open-label study will investigate first-line domvanalimab (anti-TIGIT) and zimberelimab (anti-PD-1) plus chemotherapy versus pembrolizumab plus chemotherapy in mNSCLC with no actionable gene alterations. PARTICIPANTS AND METHODS Approximately 720 participants (≥18 years old) with untreated mNSCLC and no EGFR and ALK mutations will be randomized into 3 groups (A, B, or C) in a 4:4:1 ratio and stratified by baseline PD-L1 expression (tumor cells <50% vs. ≥50%), histology (squamous vs. nonsquamous), and geographic region (East Asia vs. non-East Asia). Group A will receive domvanalimab 1200 mg plus zimberelimab 360 mg plus platinum-doublet chemotherapy (PT), group B will receive pembrolizumab 200 mg plus PT, and group C will receive zimberelimab 360 mg plus PT, every 3 weeks. Treatment will be administered until disease progression or intolerable toxicity. Dual primary endpoints are progression-free survival (by blinded independent central review [BICR]) and overall survival for group A versus B. Key secondary endpoints comprise overall response rate (by BICR), safety, and quality of life. Exploratory endpoints include efficacy and safety between groups A and C, pharmacokinetics, patient-reported outcomes, and biomarkers. CONCLUSION Enrollment in the STAR-121 study commenced on October 12, 2022, and is currently ongoing with completion planned by September 2024. The study completion is expected by December 2027.
Collapse
Affiliation(s)
- Delvys Rodriguez-Abreu
- Complejo Hospitalario Universitario Insular-Materno Infantil de Gran Canaria, Universidad de Las Palmas de Gran Canaria, Gran Canaria, Spain.
| | - Joaquim Bosch-Barrera
- Department of Medical Oncology, Catalan Institute of Oncology, Doctor Josep Trueta University Hospital; Precision Oncology Group (OncoGIR-Pro), Institut d'Investigació Biomèdica de Girona (IDIBGI); Department of Medical Sciences, Medical School, University of Girona, Girona, Spain
| | | | - Myung-Ju Ahn
- Department of Hemato-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, The Republic of Korea
| | | | - Xinwei Yu
- Gilead Sciences, Inc., Foster City, CA
| | | | | | | | | | - Martin Reck
- Lung Clinic Grosshansdorf, Airway Research Center North, German Center for Lung Research, Grosshansdorf, Germany
| |
Collapse
|
179
|
Wang X, Song J, Hu L, Ren G, Geng N, Song Z. Intrapleural perfusion hyperthermia improves the efficiency of anti‑PD1 antibody‑based therapy for lung adenocarcinoma: A case report. Oncol Lett 2024; 27:217. [PMID: 38586203 PMCID: PMC10995656 DOI: 10.3892/ol.2024.14351] [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: 09/15/2023] [Accepted: 02/12/2024] [Indexed: 04/09/2024] Open
Abstract
Chemotherapy based on intrapleural perfusion hyperthermia (IPH) can markedly improve the sensitivity of lung adenocarcinoma cells to anti-programmed cell death receptor 1 (PD1) antibody adjuvant chemotherapy and enhance the clinical response of a patient. In the present study, a unique case of a patient who failed to respond to immunotherapy combined with chemotherapy but achieved prolonged stable disease after treatment with IPH and subsequent sintilimab-based treatment, is reported. A 50-year-old Chinese female patient was admitted to a regional cancer hospital presenting with hemoptysis and persistent fever. The findings of computed tomography imaging and thoracic puncture tissue biopsy indicated a diagnosis of adenocarcinoma. The TNM and clinical stage were identified as cT2N3M0 and stage IIIB, respectively. Immunohistochemical tests showed the expression of programmed death-ligand 1 (PD-L1) with a tumor proportion score of 2%. No other classic genetic alterations were detected. Initially, sintilimab-based chemotherapy at 200 mg was administered, for three cycles from April 2020, and increased pleural effusion was observed on the left side. The best overall response (BOR) assessment of the local lesion was progressive disease. IPH combined with chemotherapy was then carried out from August to September 2020, after which the same course of sintilimab-based chemotherapy as aforementioned was provided from October 2020 to September 2023. The BOR evaluation results during the monotherapy courses were all judged as stable disease. Therefore, it was concluded that IPH can substantially improve the efficiency of anti-PD1 antibody-based therapy for lung adenocarcinoma.
Collapse
Affiliation(s)
- Xiaolei Wang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, Hebei 071000, P.R. China
| | - Jin Song
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, Hebei 071000, P.R. China
| | - Ling Hu
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, Hebei 071000, P.R. China
| | - Guanying Ren
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, Hebei 071000, P.R. China
| | - Nan Geng
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Zizheng Song
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Baoding, Hebei 071000, P.R. China
| |
Collapse
|
180
|
Raad A, Rizzo M, Appiah K, Kearns I, Hernandez L. Critical Examination of Modeling Approaches Used in Economic Evaluations of First-Line Treatments for Locally Advanced or Metastatic Non-Small Cell Lung Cancer Harboring Epidermal Growth Factor Receptor Mutations: A Systematic Literature Review. PHARMACOECONOMICS 2024; 42:527-568. [PMID: 38489077 PMCID: PMC11039500 DOI: 10.1007/s40273-024-01362-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/11/2024] [Indexed: 03/17/2024]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, with up to 32% of patients with NSCLC harboring an epidermal growth factor receptor (EGFR) mutation. NSCLC harboring an EGFR mutation has a dedicated treatment pathway, with EGFR tyrosine kinase inhibitors and platinum-based chemotherapy often being the therapy of choice. OBJECTIVE The aim of this study was to systemically review and summarize economic models of first-line treatments used for locally advanced or metastatic NSCLC harboring EGFR mutations, as well as to identify areas for improvement for future models. METHODS Literature searches were conducted via Ovid in PubMed, MEDLINE, MEDLINE In-Process, Embase, Evidence-Based Medicine Reviews: Health Technology Assessment, Evidence-Based Medicine Reviews: National Health Service Economic Evaluation Database, and EconLit. An initial search was conducted on 19 December 2022 and updated on 11 April 2023. Studies were selected according to predefined criteria using the Population, Intervention, Comparator, Outcome and Study design (PICOS) framework. RESULTS Sixty-seven articles were included in the review, representing 59 unique studies. The majority of included models were cost-utility analyses (n = 52), with the remaining studies being cost-effectiveness analyses (n = 4) and a cost-minimization analysis (n = 1). Two studies incorporated both a cost-utility and cost-minimization analysis. Although the model structure across studies was consistently reported, justification for this choice was often lacking. CONCLUSIONS Although the reporting of economic models in NSCLC harboring EGFR mutations is generally good, many of these studies lacked sufficient reporting of justification for structural choices, performing extensive sensitivity analyses and validation in economic evaluations. In resolving such gaps, the validity of future models can be increased to guide healthcare decision making in rare indications.
Collapse
Affiliation(s)
| | | | | | | | - Luis Hernandez
- Takeda Pharmaceuticals America, Inc., Lexington, MA, USA.
| |
Collapse
|
181
|
Mariniello A, Nasti TH, Chang DY, Hashimoto M, Malik S, McManus DT, Lee J, McGuire DJ, Cardenas MA, Umana P, Nicolini V, Antia R, Saha A, Buchwald Z, Kissick H, Ghorani E, Novello S, Sangiolo D, Scagliotti GV, Ramalingam SS, Ahmed R. Platinum-Based Chemotherapy Attenuates the Effector Response of CD8 T Cells to Concomitant PD-1 Blockade. Clin Cancer Res 2024; 30:1833-1845. [PMID: 37992307 PMCID: PMC11061601 DOI: 10.1158/1078-0432.ccr-23-1316] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 09/27/2023] [Accepted: 11/20/2023] [Indexed: 11/24/2023]
Abstract
PURPOSE Combination of chemotherapy with programmed cell death 1 (PD-1) blockade is a front-line treatment for lung cancer. However, it remains unknown whether and how chemotherapy affects the response of exhausted CD8 T cells to PD-1 blockade. EXPERIMENTAL DESIGN We used the well-established mouse model of T-cell exhaustion with chronic lymphocytic choriomeningitis virus (LCMV) infection to assess the effect of chemotherapy (cisplatin+pemetrexed) on T-cell response to PD-1 blockade, in the absence of the impact of chemotherapy on antigen release and presentation observed in tumor models. RESULTS When concomitantly administered with PD-1 blockade, chemotherapy affected the differentiation path of LCMV-specific CD8 T cells from stem-like to transitory effector cells, thereby reducing their expansion and production of IFNγ. After combination treatment, these restrained effector responses resulted in impaired viral control, compared with PD-1 blockade alone. The sequential combination strategy, where PD-1 blockade followed chemotherapy, proved to be superior to the concomitant combination, preserving the proliferative response of exhausted CD8 T cells to PD-1 blockade. Our findings suggest that the stem-like CD8 T cells themselves are relatively unaffected by chemotherapy partly because they are quiescent and maintained by slow self-renewal at the steady state. However, upon the proliferative burst mediated by PD-1 blockade, the accelerated differentiation and self-renewal of stem-like cells may be curbed by concomitant chemotherapy, ultimately resulting in impaired overall CD8 T-cell effector functions. CONCLUSIONS In a translational context, we provide a proof-of-concept to consider optimizing the timing of chemo-immunotherapy strategies for improved CD8 T-cell functions. See related commentary by Vignali and Luke, p. 1705.
Collapse
Affiliation(s)
- Annapaola Mariniello
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Department of Oncology, University of Torino, Turin, Italy
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Tahseen H. Nasti
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Daniel Y. Chang
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Masao Hashimoto
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Sakshi Malik
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Daniel T. McManus
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Judong Lee
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Donald J. McGuire
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Maria A. Cardenas
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia
| | - Pablo Umana
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Valeria Nicolini
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Rustom Antia
- Department of Biology, Emory University, Atlanta, Georgia
| | - Ananya Saha
- Department of Biology, Emory University, Atlanta, Georgia
| | - Zachary Buchwald
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia
| | - Hayden Kissick
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
- Department of Urology, Emory University School of Medicine, Atlanta, Georgia
| | - Ehsan Ghorani
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Cancer Immunology and Immunotherapy Unit, Imperial College London, Department of Surgery and Cancer, London, United Kingdom
| | - Silvia Novello
- Department of Oncology, University of Torino, Turin, Italy
| | - Dario Sangiolo
- Department of Oncology, University of Torino, Turin, Italy
| | | | - Suresh S. Ramalingam
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Rafi Ahmed
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
- Winship Cancer Institute, Winship Cancer Institute of Emory University, Atlanta, Georgia
| |
Collapse
|
182
|
Remon J, Auclin E, Zubiri L, Schneider S, Rodriguez-Abreu D, Minatta N, Gautschi O, Aboubakar F, Muñoz-Couselo E, Pierret T, Rothschild SI, Cortiula F, Reynolds KL, Thibault C, Gavralidis A, Blais N, Barlesi F, Planchard D, Besse BMD. Immune checkpoint blockers in solid organ transplant recipients and cancer: the INNOVATED cohort. ESMO Open 2024; 9:103004. [PMID: 38653155 PMCID: PMC11053286 DOI: 10.1016/j.esmoop.2024.103004] [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: 01/27/2024] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Patients with solid organ transplant (SOT) and solid tumors are usually excluded from clinical trials testing immune checkpoint blockers (ICB). As transplant rates are increasing, we aimed to evaluate ICB outcomes in this population, with a special focus on lung cancer. METHODS We conducted a multicenter retrospective cohort study collecting real data of ICB use in patients with SOT and solid tumors. Clinical data and treatment outcomes were assessed by using retrospective medical chart reviews in every participating center. Study endpoints were: overall response rate (ORR), 6-month progression-free survival (PFS), and grade ≥3 immune-related adverse events. RESULTS From August 2016 to October 2022, 31 patients with SOT (98% kidney) and solid tumors were identified (36.0% lung cancer, 19.4% melanoma, 13.0% genitourinary cancer, 6.5% gastrointestinal cancer). Programmed death-ligand 1 expression was positive in 29% of tumors. Median age was 61 years, 69% were males, and 71% received ICB as first-line treatment. In the whole cohort the ORR was 45.2%, with a 6-month PFS of 56.8%. In the lung cancer cohort, the ORR was 45.5%, with a 6-month PFS of 32.7%, and median overall survival of 4.6 months. The grade 3 immune-related adverse events rate leading to ICB discontinuation was 12.9%. Allograft rejection rate was 25.8%, and risk of rejection was similar regardless of the type of ICB strategy (monotherapy or combination, 28% versus 33%, P = 1.0) or response to ICB treatment. CONCLUSIONS ICB could be considered a feasible option for SOT recipients with some advanced solid malignancies and no alternative therapeutic options. Due to the risk of allograft rejection, multidisciplinary teams should be involved before ICB therapy.
Collapse
Affiliation(s)
- J Remon
- Paris-Saclay University, Department of Cancer Medicine, Gustave Roussy, Villejuif.
| | - E Auclin
- Department of Cancer Medicine, Hôpital Européen Georges-Pompidou, Paris, France
| | - L Zubiri
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, USA
| | - S Schneider
- Department Pneumology, Hôpital de Bayonne, Bayonne, France
| | - D Rodriguez-Abreu
- Medical Oncology Department, Complejo Hospitalario Universitario Insular-Materno Infantil de Gran Canaria, Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - N Minatta
- Department of Oncology Hospital Italiano Buenos Aires, Buenos Aires, Argentina
| | - O Gautschi
- Department of Cancer Medicine, University of Berne and Cantonal Hospital of Lucerne, Lucerne, Switzerland
| | - F Aboubakar
- Department of Pneumology, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - E Muñoz-Couselo
- Department of Oncology, Hospital Vall d'Hebron de Barcelona, VHIO Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - T Pierret
- Department of Pneumology, CHU Grenoble Alpes, Grenoble, France
| | - S I Rothschild
- Medical Oncology Department, University Hospital Basel, Basel; Division Oncology/Hematology, Department of Medicine, Cantonal Hospital Baden, Baden, Switzerland
| | - F Cortiula
- Department of Oncology, University Hospital of Udine, Udine, Italy
| | - K L Reynolds
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, USA
| | - C Thibault
- Department of Cancer Medicine, Hôpital Européen Georges-Pompidou, Paris, France
| | - A Gavralidis
- Massachusetts General Hospital Cancer Center, Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston, USA; Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Boston; Salem Hospital, Salem, USA
| | - N Blais
- Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, Canada
| | - F Barlesi
- Paris-Saclay University, Department of Cancer Medicine, Gustave Roussy, Villejuif
| | - D Planchard
- Paris-Saclay University, Department of Cancer Medicine, Gustave Roussy, Villejuif
| | - B M D Besse
- Paris-Saclay University, Department of Cancer Medicine, Gustave Roussy, Villejuif
| |
Collapse
|
183
|
Torres-Jiménez J, Espinar JB, de Cabo HB, Berjaga MZ, Esteban-Villarrubia J, Fraile JZ, Paz-Ares L. Targeting KRAS G12C in Non-Small-Cell Lung Cancer: Current Standards and Developments. Drugs 2024; 84:527-548. [PMID: 38625662 DOI: 10.1007/s40265-024-02030-7] [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: 03/27/2024] [Indexed: 04/17/2024]
Abstract
Among the most common molecular alterations detected in non-small-cell lung cancer (NSCLC) are mutations in Kristen Rat Sarcoma viral oncogene homolog (KRAS). KRAS mutant NSCLC is a heterogenous group of diseases, different from other oncogene-driven tumors in terms of biology and response to therapies. Despite efforts to develop drugs aimed at inhibiting KRAS or its signaling pathways, KRAS had remained undruggable for decades. The discovery of a small pocket in the binding switch II region of KRASG12C has revolutionized the treatment of KRASG12C-mutated NSCLC patients. Sotorasib and adagrasib, direct KRASG12C inhibitors, have been approved by the US Food and Drug Administration (FDA) and other regulatory agencies for patients with previously treated KRASG12C-mutated NSCLC, and these advances have become practice changing. However, first-line treatment in KRASG12C-mutated NSCLC does not differ from NSCLC without actionable driver genomic alterations. Treatment with KRASG12C inhibitors is not curative and patients develop progressive disease, so understanding associated mechanisms of drug resistance is key. New KRASG12C inhibitors and several combination therapy strategies, including with immune checkpoint inhibitors, are being studied in clinical trials. The aim of this review is to explore the clinical impact of KRAS, and outline different treatment approaches, focusing on the novel treatment of KRASG12C-mutated NSCLC.
Collapse
Affiliation(s)
- Javier Torres-Jiménez
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Avda de Córdoba s/n, 28041, Madrid, Spain.
| | - Javier Baena Espinar
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Avda de Córdoba s/n, 28041, Madrid, Spain
| | - Helena Bote de Cabo
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Avda de Córdoba s/n, 28041, Madrid, Spain
| | - María Zurera Berjaga
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Avda de Córdoba s/n, 28041, Madrid, Spain
| | - Jorge Esteban-Villarrubia
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Avda de Córdoba s/n, 28041, Madrid, Spain
| | - Jon Zugazagoitia Fraile
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Avda de Córdoba s/n, 28041, Madrid, Spain
- Lung Cancer Group, Clinical Research Program, CNIO (Centro Nacional de Investigaciones Oncológicas) and Instituto de Investigación i+12, Madrid, Spain
| | - Luis Paz-Ares
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Avda de Córdoba s/n, 28041, Madrid, Spain
- Lung Cancer Group, Clinical Research Program, CNIO (Centro Nacional de Investigaciones Oncológicas) and Instituto de Investigación i+12, Madrid, Spain
| |
Collapse
|
184
|
Bischoff P, Reck M, Overbeck T, Christopoulos P, Rittmeyer A, Lüders H, Kollmeier J, Kulhavy J, Kemper M, Reinmuth N, Röper J, Janning M, Sommer L, Aguinarte L, Koch M, Wiesweg M, Wesseler C, Waller CF, Kauffmann-Guerrero D, Stenzinger A, Stephan-Falkenau S, Trautmann M, Lassmann S, Tiemann M, Klauschen F, Sebastian M, Griesinger F, Wolf J, Loges S, Frost N. Outcome of First-Line Treatment With Pembrolizumab According to KRAS/TP53 Mutational Status for Nonsquamous Programmed Death-Ligand 1-High (≥50%) NSCLC in the German National Network Genomic Medicine Lung Cancer. J Thorac Oncol 2024; 19:803-817. [PMID: 38096950 DOI: 10.1016/j.jtho.2023.12.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 01/18/2024]
Abstract
INTRODUCTION Programmed death-ligand 1 expression currently represents the only validated predictive biomarker for immune checkpoint inhibition in metastatic NSCLC in the clinical routine, but it has limited value in distinguishing responses. Assessment of KRAS and TP53 mutations (mut) as surrogate for an immunosupportive tumor microenvironment (TME) might help to close this gap. METHODS A total of 696 consecutive patients with programmed death-ligand 1-high (≥50%), nonsquamous NSCLC, having received molecular testing within the German National Network Genomic Medicine Lung Cancer between 2017 and 2020, with Eastern Cooperative Oncology Group performance status less than or equal to 1 and pembrolizumab as first-line palliative treatment, were included into this retrospective cohort analysis. Treatment efficacy and outcome according to KRAS/TP53 status were correlated with TME composition and gene expression analysis of The Cancer Genome Atlas lung adenocarcinoma cohort. RESULTS Proportion of KRASmut and TP53mut was 53% (G12C 25%, non-G12C 28%) and 51%, respectively. In KRASmut patients, TP53 comutations increased response rates (G12C: 69.7% versus 46.5% [TP53mut versus wild-type (wt)], p = 0.004; non-G12C: 55.4% versus 39.5%, p = 0.03), progression-free survival (G12C: hazard ratio [HR] = 0.59, p = 0.009, non-G12C: HR = 0.7, p = 0.047), and overall survival (G12C: HR = 0.72, p = 0.16, non-G12C: HR = 0.56, p = 0.002), whereas no differences were observed in KRASwt patients. After a median follow-up of 41 months, G12C/TP53mut patients experienced the longest progression-free survival and overall survival (33.7 and 65.3 mo), which correlated with high tumor-infiltrating lymphocyte densities in the TME and up-regulation of interferon gamma target genes. Proinflammatory pathways according to TP53 status (mut versus wt) were less enhanced and not different in non-G12C and KRASwt, respectively. CONCLUSIONS G12C/TP53 comutations identify a subset of patients with a very favorable long-term survival with immune checkpoint inhibitor monotherapy, mediated by highly active interferon gamma signaling in a proinflammatory TME.
Collapse
Affiliation(s)
- Philip Bischoff
- Institute of Pathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany; BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany; German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Reck
- Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany
| | - Tobias Overbeck
- Department of Haematology and Medical Oncology, University Medical Center Göttingen and Lungentumorzentrum Universität Göttingen, Göttingen, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Achim Rittmeyer
- Department of Thoracic Oncology, LKI Lungenfachklinik Immenhausen, Immenhausen, Germany
| | - Heike Lüders
- Klinik für Pneumologie-Evangelische Lungenklinik Berlin Buch, Berlin, Germany
| | - Jens Kollmeier
- Helios Klinikum Emil von Behring, Lungenklinik Heckeshorn, Berlin, Germany; Berlin Lung Institute, Berlin, Germany
| | - Jonas Kulhavy
- Translational Oncology/Early Clinical Trial Unit (ECTU), Comprehensive Cancer Center Mainfranken and Bavarian Cancer Research Center (BZKF), University Hospital Wuerzburg, Wuerzburg, Germany
| | - Marcel Kemper
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | - Niels Reinmuth
- Asklepios Lung Clinic, member of the German Center for Lung Research (DZL), Munich-Gauting, Germany
| | - Julia Röper
- Department of Hematology and Oncology, Pius-Hospital, University Dept. of Internal Medicine-Oncology, Oldenburg, Germany
| | - Melanie Janning
- DKFZ-Hector Cancer Institute and Department of Personalized Oncology at the University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Linna Sommer
- Department of Thoracic Oncology, Carl-Gustav-Carus Dresden University Hospital, Dresden, Germany
| | - Lukas Aguinarte
- Hematology/Oncology, Department of Medicine II, University Hospital Frankfurt, Frankfurt, Germany
| | - Myriam Koch
- University Hospital Regensburg, Department of Internal Medicine 2, Regensburg, Germany
| | - Marcel Wiesweg
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Claas Wesseler
- Department of Thoracic Oncology, Asklepios Klinikum Harburg, Hamburg, Germany
| | - Cornelius F Waller
- Department of Haematology, Oncology and Stem Cell Transplantation, University Medical Centre Freiburg and Faculty of Medicine, Freiburg, Germany
| | - Diego Kauffmann-Guerrero
- Department of Medicine V, University Hospital, LMU Munich, Member of the German Center for Lung Research (DZL-CPCM), Munich, Germany
| | | | | | - Marcel Trautmann
- University of Münster, Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Silke Lassmann
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Frederick Klauschen
- Institute of Pathology, Ludwig-Maximilians-University, Munich, Germany; Berlin Institute for the Foundation of Learning and Data (BIFOLD) and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Martin Sebastian
- Hematology/Oncology, Department of Medicine II, University Hospital Frankfurt, Frankfurt, Germany
| | - Frank Griesinger
- Department of Hematology and Oncology, Pius-Hospital, University Dept. of Internal Medicine-Oncology, Oldenburg, Germany
| | - Jürgen Wolf
- Department I of Internal Medicine, Center for Integrated Oncology (CIO), University Hospital of Cologne, Cologne, Germany
| | - Sonja Loges
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Nikolaj Frost
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin (Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Berlin, Germany.
| |
Collapse
|
185
|
Leonetti A, Perrone F, Puntoni M, Maglietta G, Bordi P, Bria E, Vita E, Gelsomino F, De Giglio A, Gelibter A, Siringo M, Mazzoni F, Caliman E, Genova C, Bertolini F, Guaitoli G, Passiglia F, Delcuratolo MD, Montrone M, Cerea G, Pasello G, Roca E, Belluomini L, Cecere FL, Guida A, Manzo A, Adamo V, Rastelli F, Bulotta A, Citarella F, Toschi L, Zoratto F, Cortinovis DL, Berardi R, Follador A, Carta A, Camerini A, Salerno F, Silva RR, Baldini E, Cortellini A, Brighenti M, Santoni M, Malorgio F, Caminiti C, Tiseo M. Real-world outcomes of Italian patients with advanced non-squamous lung cancer treated with first-line pembrolizumab plus platinum-pemetrexed. Eur J Cancer 2024; 202:114006. [PMID: 38489861 DOI: 10.1016/j.ejca.2024.114006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 03/03/2024] [Indexed: 03/17/2024]
Abstract
PURPOSE The aim of this multi-center, retrospective/prospective cohort observational study was to evaluate outcomes in routine clinical practice of first-line chemo-immunotherapy with cis/carboplatin, pemetrexed and pembrolizumab in patients with advanced non-squamous non-small cell lung cancer (NSCLC) in 33 Italian centers. METHODS The outcome measure was to evaluate overall survival (OS) in a real-world patient population. Secondary endpoints were: progression-free survival (PFS), objective response rate (ORR), duration of response (DoR) and incidence of treatment-related adverse events (AEs). RESULTS 1068 patients were enrolled at the time of data cut-off (January 31st, 2023), and 812 (76.0%) belonged to the retrospective cohort. Median age was 66 years (27-85), ECOG PS was ≥ 2 in 91 (8.6%) patients; 254 (23.8%) patients had brain metastases at baseline; 38 (3.6%) patients had tumor with PD-L1 expression ≥ 50%. After a median follow-up of 17.0 months (95% CI, 16.1-17.9), median OS was 16.1 months (95% CI, 14.4-18.8) and PFS was 9.9 months (95% CI, 8.8-11.2). Median DoR (n = 493) was 14.7 months (95% CI, 13.6-17.1). ORR was 43.4% (95% CI, 40.4-46.4). Any-grade AEs occurred in 636 (59.6%) patients and grade ≥ 3 in 253 (23.7%) patients. Most common grade ≥ 3 AEs were neutropenia (6.3%) and anemia (6.3%). CONCLUSIONS First-line chemo-immunotherapy was effective and tolerable in this large, real-world Italian study of patients with advanced non-squamous NSCLC. Our results were in line with the KEYNOTE-189 registration study, also considering the low number of PD-L1 ≥ 50% patients included in our study.
Collapse
Affiliation(s)
| | - Fabiana Perrone
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy.
| | - Matteo Puntoni
- Clinical & Epidemiological Research Unit, University Hospital of Parma, Parma, Italy
| | - Giuseppe Maglietta
- Clinical & Epidemiological Research Unit, University Hospital of Parma, Parma, Italy
| | - Paola Bordi
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Emilio Bria
- UOSD Oncologia Toraco-Polmonare, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Medical Oncology, Department of Traslational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Emanuele Vita
- UOSD Oncologia Toraco-Polmonare, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Medical Oncology, Department of Traslational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Francesco Gelsomino
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrea De Giglio
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Alain Gelibter
- Medical Oncology (B), Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Marco Siringo
- Medical Oncology (B), Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | | | - Enrico Caliman
- Medical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Carlo Genova
- Academic Oncology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy; Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy
| | - Federica Bertolini
- Division of Medical Oncology, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Giorgia Guaitoli
- Division of Medical Oncology, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Francesco Passiglia
- Department of Oncology, University of Turin, AOU San Luigi Gonzaga, Orbassano, Turin, Italy
| | | | - Michele Montrone
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Giulio Cerea
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giulia Pasello
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padua, Italy; Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Elisa Roca
- Thoracic Oncology - Lung Unit, Pederzoli Hospital, Peschiera del Garda, Italy
| | - Lorenzo Belluomini
- Medical Oncology, Department of Medicine, University of Verona Hospital Trust, Verona, Italy
| | | | - Annalisa Guida
- Department of Medical Oncology, St. Mary's Hospital, Terni, Italy
| | - Anna Manzo
- Thoracic Medical Oncology, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy
| | - Vincenzo Adamo
- Medical Oncology Unit, Azienda Ospedaliera Papardo, Messina, Italy
| | - Francesca Rastelli
- Medical Oncology, AST (Azienda Sanitaria Territoriale) of Ascoli Piceno, Ascoli Piceno, Italy
| | - Alessandra Bulotta
- Department of Oncology, Istituto di Ricerca a Carattere Scientifico (IRCCS) San Raffaele Hospital, Milan, Italy
| | - Fabrizio Citarella
- Department of Medical Oncology, Campus Bio-Medico University, Rome, Italy
| | - Luca Toschi
- IRCCS Humanitas Clinical and Research Center - Humanitas Cancer Center, Rozzano, Milan, Italy
| | | | - Diego Luigi Cortinovis
- SC Medical Oncology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy; Medicine and Surgery Department, University of Milano Bicocca, Milan, Italy
| | - Rossana Berardi
- Oncology Clinic, Università Politecnica Delle Marche, Ospedali Riuniti Di Ancona, Ancona, Italy
| | - Alessandro Follador
- Medical Oncology Unit San Daniele - Tolmezzo, ASUFC Azienda Sanitaria Universitaria Friuli Centrale, Italy
| | - Annamaria Carta
- Pathology and Oncology Unit, Businco Oncological Hospital, Cagliari, Italy
| | - Andrea Camerini
- Medical Oncology, Versilia Hospital, Azienda USL Toscana Nord Ovest, Lido di Camaiore, Italy
| | - Flavio Salerno
- Department of Oncology, University of Turin, Ordine Mauriziano Hospital, Turin, Italy
| | - Rosa Rita Silva
- Department of Oncology, ASUR Marche, Area Vasta 2, Fabriano, Italy
| | | | - Alessio Cortellini
- Operative Research Unit of Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128, Roma, Italy; Department of Surgery and Cancer, Hammersmith Hospital Campus, Imperial College London, London, UK
| | | | | | | | - Caterina Caminiti
- Clinical & Epidemiological Research Unit, University Hospital of Parma, Parma, Italy
| | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy; Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
186
|
Werner R, Steinmann N, Decaluwe H, Date H, De Ruysscher D, Opitz I. Complex situations in lung cancer: multifocal disease, oligoprogression and oligorecurrence. Eur Respir Rev 2024; 33:230200. [PMID: 38811031 PMCID: PMC11134198 DOI: 10.1183/16000617.0200-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 02/23/2024] [Indexed: 05/31/2024] Open
Abstract
With the emergence of lung cancer screening programmes and newly detected localised and multifocal disease, novel treatment compounds and multimodal treatment approaches, the treatment landscape of non-small cell lung cancer is becoming increasingly complex. In parallel, in-depth molecular analyses and clonality studies are revealing more information about tumorigenesis, potential therapeutical targets and the origin of lesions. All can play an important role in cases with multifocal disease, oligoprogression and oligorecurrence. In multifocal disease, it is essential to understand the relatedness of separate lesions for treatment decisions, because this information distinguishes separate early-stage tumours from locally advanced or metastatic cancer. Clonality studies suggest that a majority of same-histology lesions represent multiple primary tumours. With the current standard of systemic treatment, oligoprogression after an initial treatment response is a common scenario. In this state of induced oligoprogressive disease, local ablative therapy by either surgery or radiotherapy is becoming increasingly important. Another scenario involves the emergence of a limited number of metastases after radical treatment of the primary tumour, referred to as oligorecurrence, for which the use of local ablative therapy holds promise in improving survival. Our review addresses these complex situations in lung cancer by discussing current evidence, knowledge gaps and treatment recommendations.
Collapse
Affiliation(s)
- Raphael Werner
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Nina Steinmann
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Herbert Decaluwe
- Department of Thoracovascular Surgery, Ziekenhuis Oost-Limburg, Genk, Belgium
| | - Hiroshi Date
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Dirk De Ruysscher
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Radiation Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Isabelle Opitz
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
187
|
Rossi S, Masini S, Finocchiaro G, Lorenzi E, Toschi L, Santoro A. Retreatment with Immune Checkpoint Inhibitors in the New Scenario of Immunotherapy in Non-Small Cell Lung Cancer. Cancers (Basel) 2024; 16:1683. [PMID: 38730635 PMCID: PMC11083698 DOI: 10.3390/cancers16091683] [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: 03/28/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
The advent of immunotherapy has transformed the treatment paradigm for metastatic non-small cell lung cancer (NSCLC). In the past few years, several studies have investigated the potential role of immune checkpoint inhibitors (ICIs) in resectable and unresectable locally advanced disease, achieving remarkable results that led to their approval in clinical practice. However, there is limited evidence on immunotherapy rechallenge after recurrence, with the majority of available knowledge coming from retrospective studies which involve heavily pretreated patients with advanced NSCLC. The recent introduction in the curative setting and the potential regulatory restrictions raise questions about the optimal choice of first-line and subsequent therapies for patients with systemic relapse. The role of immunotherapy readministration in this new scenario needs to be clarified, as well as the identification of patients for whom it is more appropriate, including clinical characteristics, duration of response, switching to other ICIs, reasons for discontinuation and immune-related toxicity. Here, we review literature on rechallenge with immunotherapy, including efficacy, safety profile and potential predictive factors of response.
Collapse
Affiliation(s)
- Sabrina Rossi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, Rozzano, 20089 Milan, Italy; (S.R.); (G.F.); (E.L.); (L.T.); (A.S.)
| | - Silvia Masini
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, Rozzano, 20089 Milan, Italy; (S.R.); (G.F.); (E.L.); (L.T.); (A.S.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
| | - Giovanna Finocchiaro
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, Rozzano, 20089 Milan, Italy; (S.R.); (G.F.); (E.L.); (L.T.); (A.S.)
| | - Elena Lorenzi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, Rozzano, 20089 Milan, Italy; (S.R.); (G.F.); (E.L.); (L.T.); (A.S.)
| | - Luca Toschi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, Rozzano, 20089 Milan, Italy; (S.R.); (G.F.); (E.L.); (L.T.); (A.S.)
| | - Armando Santoro
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, Rozzano, 20089 Milan, Italy; (S.R.); (G.F.); (E.L.); (L.T.); (A.S.)
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
| |
Collapse
|
188
|
Anguera G, Mulet M, Zamora C, Osuna-Gómez R, Barba A, Sullivan I, Serra-López J, Cantó E, Vidal S, Majem M. Potential Role of Circulating PD-L1 + Leukocytes as a Predictor of Response to Anti-PD-(L)1 Therapy in NSCLC Patients. Biomedicines 2024; 12:958. [PMID: 38790920 PMCID: PMC11117542 DOI: 10.3390/biomedicines12050958] [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: 03/06/2024] [Revised: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
PD-(L)1 inhibitors are part of the treatment strategy for non-small cell lung cancer (NSCLC) although its efficacy is limited to certain patients. Our study aimed to identify patients who might benefit from anti-PD-(L)1 inhibitors by analyzing the PD-L1 expression on circulating leukocytes and its evolution during treatment. One hundred thirteen NSCLC patients, according to their radiological response after 10-12 weeks of treatment, were classified into responders, stable, and progressive disease. Percentages of circulating PD-L1+ leukocytes, PD-L1+ platelets (PLTs), and leukocyte-PLT complexes were assessed using flow cytometry, and plasma concentrations of soluble immunomodulatory factors were quantified by ELISA. Responders exhibited significantly higher pre-treatment percentages of PD-L1+ neutrophils, PD-L1+ CD14+ cells, and PD-L1+ PLTs than progressors. The percentages of these populations decreased in responders post-treatment, contrasting with stables and progressors. PLTs notably contributed to PD-L1 expression in CD14+ cells and neutrophils. Plasma cytokine analysis revealed baseline differences only in IL-17 concentration among groups, whereas network analyses highlighted distinct association patterns between plasma molecules and PD-L1+ leukocytes after 10-12 weeks of treatment. Our findings suggest that pre-treatment assessment of circulating PD-L1+ neutrophils, PD-L1+ CD14+ cells, and PD-L1+ PLTs may be helpful in identifying NSCLC patients who are potential candidates for anti-PD-(L)1 therapy.
Collapse
Affiliation(s)
- Georgia Anguera
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (G.A.); (A.B.); (I.S.); (J.S.-L.); (M.M.)
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Maria Mulet
- Group of Inflammatory Diseases, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (C.Z.); (R.O.-G.); (E.C.); (S.V.)
| | - Carlos Zamora
- Group of Inflammatory Diseases, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (C.Z.); (R.O.-G.); (E.C.); (S.V.)
| | - Rubén Osuna-Gómez
- Group of Inflammatory Diseases, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (C.Z.); (R.O.-G.); (E.C.); (S.V.)
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Andrés Barba
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (G.A.); (A.B.); (I.S.); (J.S.-L.); (M.M.)
| | - Ivana Sullivan
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (G.A.); (A.B.); (I.S.); (J.S.-L.); (M.M.)
| | - Jorgina Serra-López
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (G.A.); (A.B.); (I.S.); (J.S.-L.); (M.M.)
| | - Elisabet Cantó
- Group of Inflammatory Diseases, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (C.Z.); (R.O.-G.); (E.C.); (S.V.)
| | - Silvia Vidal
- Group of Inflammatory Diseases, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (C.Z.); (R.O.-G.); (E.C.); (S.V.)
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Margarita Majem
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (G.A.); (A.B.); (I.S.); (J.S.-L.); (M.M.)
| |
Collapse
|
189
|
Orosz Z, Kovács Á. The role of chemoradiotherapy and immunotherapy in stage III NSCLC. Pathol Oncol Res 2024; 30:1611716. [PMID: 38706775 PMCID: PMC11066192 DOI: 10.3389/pore.2024.1611716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024]
Abstract
Locally advanced non-small lung cancer encompasses a diverse range of tumors. In the last few years, the treatment of stage III unresectable non-small lung cancer has evolved significantly. The PACIFIC trial opened a new therapeutic era in the treatment of locally advanced NSCLC, establishing durvalumab consolidation therapy as the new standard of care worldwide. A careful evaluation of this type of lung cancer and a discussion of the management of these patients within a multidisciplinary team represents a crucial step in defining the best treatment strategy for each patient. For unresectable stage III NSCLC, definitive concurrent chemoradiotherapy (CCRT) was historically recommended as a treatment with a 5-year survival rate ranging from 20% to 30%. The PACIFIC study conducted in 2017 compared the use of chemoradiotherapy and maintenance therapy with the anti-PD-L1 monoclonal antibody durvalumab to a placebo in patients with locally advanced NSCLC who had not experienced disease progression. The study was prospective, randomized, and phase III. The administration of this medication in patients with locally advanced non-small cell lung cancer (NSCLC) has demonstrated a notable improvement in overall survival. Multiple clinical trials are currently exploring various immune checkpoint inhibition regimens to enhance the treatment efficacy in patients with stage III cancer. Our goal is to offer an up-to-date summary of the planned clinical trials for treatment options, focusing on the significant obstacles and prospects in the post-PACIFIC era.
Collapse
Affiliation(s)
- Zsuzsanna Orosz
- Department of Pulmonology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Árpád Kovács
- Department of Oncoradiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
190
|
Stares M, Brown LR, Abhi D, Phillips I. Prognostic Biomarkers of Systemic Inflammation in Non-Small Cell Lung Cancer: A Narrative Review of Challenges and Opportunities. Cancers (Basel) 2024; 16:1508. [PMID: 38672590 PMCID: PMC11048253 DOI: 10.3390/cancers16081508] [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: 03/04/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is a common malignancy and is associated with poor survival outcomes. Biomarkers of systemic inflammation derived from blood tests collected as part of routine clinical care offer prognostic information for patients with NSCLC that may assist clinical decision making. They are an attractive tool, as they are inexpensive, easily measured, and reproducible in a variety of healthcare settings. Despite the wealth of evidence available to support them, these inflammatory biomarkers are not yet routinely used in clinical practice. In this narrative review, the key inflammatory indices reported in the literature and their prognostic significance in NSCLC are described. Key challenges limiting their clinical application are highlighted, including the need to define the optimal biomarker of systemic inflammation, a lack of understanding of the systemic inflammatory landscape of NSCLC as a heterogenous disease, and the lack of clinical relevance in reported outcomes. These challenges may be overcome with standardised recording and reporting of inflammatory biomarkers, clinicopathological factors, and survival outcomes. This will require a collaborative approach, to which this field of research lends itself. This work may be aided by the rise of data-driven research, including the potential to utilise modern electronic patient records and advanced data-analysis techniques.
Collapse
Affiliation(s)
- Mark Stares
- Edinburgh Cancer Centre, NHS Lothian, Edinburgh EH4 2XU, UK
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh EH4 2XR, UK
| | - Leo R. Brown
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh EH4 2XR, UK
| | - Dhruv Abhi
- Edinburgh Cancer Centre, NHS Lothian, Edinburgh EH4 2XU, UK
| | - Iain Phillips
- Edinburgh Cancer Centre, NHS Lothian, Edinburgh EH4 2XU, UK
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh EH4 2XR, UK
| |
Collapse
|
191
|
Hulo P, Deshayes S, Fresquet J, Chéné AL, Blandin S, Boisgerault N, Fonteneau JF, Treps L, Denis MG, Bennouna J, Fradin D, Pons-Tostivint E, Blanquart C. Use of non-small cell lung cancer multicellular tumor spheroids to study the impact of chemotherapy. Respir Res 2024; 25:156. [PMID: 38581044 PMCID: PMC10998296 DOI: 10.1186/s12931-024-02791-5] [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: 10/11/2023] [Accepted: 03/25/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Lung cancers represent the main cause of cancer related-death worldwide. Recently, immunotherapy alone or in combination with chemotherapy has deeply impacted the therapeutic care leading to an improved overall survival. However, relapse will finally occur, with no efficient second line treatment so far. New therapies development based on the comprehension of resistance mechanisms is necessary. However, the difficulties to obtain tumor samples before and after first line treatment hamper to clearly understand the consequence of these molecules on tumor cells and also to identify adapted second line therapies. METHODS To overcome this difficulty, we developed multicellular tumor spheroids (MCTS) using characterized Non-Small Cell Lung Cancer (NSCLC) cell lines, monocytes from healthy donors and fibroblasts. MCTS were treated with carboplatin-paclitaxel or -gemcitabine combinations according to clinical administration schedules. The treatments impact was studied using cell viability assay, histological analyses, 3'RNA sequencing, real-time PCR, flow cytometry and confocal microscopy. RESULTS We showed that treatments induced a decrease in cell viability and strong modifications in the transcriptomic profile notably at the level of pathways involved in DNA damage repair and cell cycle. Interestingly, we also observed a modification of genes expression considered as hallmarks of response to immune check point inhibitors and immunogenicity, particularly an increase in CD274 gene expression, coding for PD-L1. This result was validated at the protein level and shown to be restricted to tumor cells on MCTS containing fibroblasts and macrophages. This increase was also observed in an additional cell line, expressing low basal CD274 level. CONCLUSIONS This study shows that MCTS are interesting models to study the impact of first line therapies using conditions close to clinical practice and also to identify more adapted second line or concomitant therapies for lung cancer treatment.
Collapse
Affiliation(s)
- Pauline Hulo
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, Nantes, CRCI2NA, F- 44000, France
- Medical oncology, Nantes Université, CHU Nantes, Nantes, F-44000, France
| | - Sophie Deshayes
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, Nantes, CRCI2NA, F- 44000, France
| | - Judith Fresquet
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, Nantes, CRCI2NA, F- 44000, France
| | - Anne-Laure Chéné
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, Nantes, CRCI2NA, F- 44000, France
- Service de pneumologie, L'institut du thorax, Hôpital Guillaume et René Laennec, CHU Nantes, Nantes, France
| | - Stéphanie Blandin
- Nantes Université, CHU Nantes, CNRS, Inserm, BioCore, US16, SFR Bonamy, Nantes, F-44000, France
| | - Nicolas Boisgerault
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, Nantes, CRCI2NA, F- 44000, France
| | - Jean-François Fonteneau
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, Nantes, CRCI2NA, F- 44000, France
| | - Lucas Treps
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, Nantes, CRCI2NA, F- 44000, France
| | - Marc G Denis
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, Nantes, CRCI2NA, F- 44000, France
- Department of Biochemistry, Nantes Université, CHU Nantes, Nantes, F-44000, France
| | - Jaafar Bennouna
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, Nantes, CRCI2NA, F- 44000, France
- Medical oncology, Nantes Université, CHU Nantes, Nantes, F-44000, France
| | - Delphine Fradin
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, Nantes, CRCI2NA, F- 44000, France
| | - Elvire Pons-Tostivint
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, Nantes, CRCI2NA, F- 44000, France.
- Medical oncology, Nantes Université, CHU Nantes, Nantes, F-44000, France.
| | - Christophe Blanquart
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, Nantes, CRCI2NA, F- 44000, France.
| |
Collapse
|
192
|
Mfumbilwa ZA, Simons MJHG, Ramaekers B, Retèl VP, Mankor JM, Groen HJM, Aerts JGJV, Joore M, Wilschut JA, Coupé VMH. Exploring the Cost Effectiveness of a Whole-Genome Sequencing-Based Biomarker for Treatment Selection in Patients with Advanced Lung Cancer Ineligible for Targeted Therapy. PHARMACOECONOMICS 2024; 42:419-434. [PMID: 38194023 PMCID: PMC10937799 DOI: 10.1007/s40273-023-01344-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/06/2023] [Indexed: 01/10/2024]
Abstract
OBJECTIVE We aimed to perform an early cost-effectiveness analysis of using a whole-genome sequencing-based tumor mutation burden (WGS-TMB), instead of programmed death-ligand 1 (PD-L1), for immunotherapy treatment selection in patients with non-squamous advanced/metastatic non-small cell lung cancer ineligible for targeted therapy, from a Dutch healthcare perspective. METHODS A decision-model simulating individual patients with metastatic non-small cell lung cancer was used to evaluate diagnostic strategies to select first-line immunotherapy only or the immunotherapy plus chemotherapy combination. Treatment was selected using PD-L1 [A, current practice], WGS-TMB [B], and both PD-L1 and WGS-TMB [C]. Strategies D, E, and F take into account a patient's disease burden, in addition to PD-L1, WGS-TMB, and both PD-L1 and WGS-TMB, respectively. Disease burden was defined as a fast-growing tumor, a high number of metastases, and/or weight loss. A threshold of 10 mutations per mega-base was used to classify patients into TMB-high and TMB-low groups. Outcomes were discounted quality-adjusted life-years (QALYs) and healthcare costs measured from the start of first-line treatment to death. Healthcare costs includes drug acquisition, follow-up costs, and molecular diagnostic tests (i.e., standard diagnostic techniques and/or WGS for strategies involving TMB). Results were reported using the net monetary benefit at a willingness-to-pay threshold of €80,000/QALY. Additional scenario and threshold analyses were performed. RESULTS Strategy B had the lowest QALYs (1.84) and lowest healthcare costs (€120,800). The highest QALYs and healthcare costs were 2.00 and €140,400 in strategy F. In the base-case analysis, strategy A was cost effective with the highest net monetary benefit (€27,300), followed by strategy B (€26,700). Strategy B was cost effective when the cost of WGS testing was decreased by at least 24% or when immunotherapy results in an additional 0.5 year of life gained or more for TMB high compared with TMB low. Strategies C and F, which combined TMB and PD-L1 had the highest net monetary benefit (≥ €76,900) when the cost of WGS testing, immunotherapy, and chemotherapy acquisition were simultaneously reduced by at least 47%, 39%, and 43%, respectively. Furthermore, strategy C resulted in the highest net monetary benefit (≥ €39,900) in a scenario where patients with both PD-L1 low and TMB low were treated with chemotherapy instead of immunotherapy plus chemotherapy. CONCLUSIONS The use of WGS-TMB is not cost effective compared to PD-L1 for immunotherapy treatment selection in non-squamous metastatic non-small cell lung cancer in the Netherlands. WGS-TMB could become cost effective provided there is a reduction in the cost of WGS testing or there is an increase in the predictive value of WGS-TMB for immunotherapy effectiveness. Alternatively, a combination strategy of PD-L1 testing with WGS-TMB would be cost effective if used to support the choice to withhold immunotherapy in patients with a low expected benefit of immunotherapy.
Collapse
Affiliation(s)
- Zakile A Mfumbilwa
- Department of Epidemiology and Data Science, Disease Modelling and Health Care Evaluation, Amsterdam UMC, Location Vrije Universiteit Amsterdam, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
- Amsterdam Public Health, Methodology, Amsterdam, The Netherlands
- Department of Mathematics and Statistics, Sokoine University of Agriculture, Morogoro, Tanzania
| | - Martijn J H G Simons
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - Bram Ramaekers
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - Valesca P Retèl
- Department of Health Technology and Services Research, University of Twente, Enschede, The Netherlands
| | - Joanne M Mankor
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Harry J M Groen
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Manuela Joore
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht University, Maastricht, The Netherlands
| | - Janneke A Wilschut
- Department of Epidemiology and Data Science, Disease Modelling and Health Care Evaluation, Amsterdam UMC, Location Vrije Universiteit Amsterdam, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
- Amsterdam Public Health, Methodology, Amsterdam, The Netherlands
| | - Veerle M H Coupé
- Department of Epidemiology and Data Science, Disease Modelling and Health Care Evaluation, Amsterdam UMC, Location Vrije Universiteit Amsterdam, PO Box 7057, 1007 MB, Amsterdam, The Netherlands.
- Amsterdam Public Health, Methodology, Amsterdam, The Netherlands.
| |
Collapse
|
193
|
Amakusa Y, Suzuki T, Hikosaka Y, Takemura M, Oguri T. Successful treatment of simultaneous malignant pleural mesothelioma and pulmonary adenocarcinoma: A case report. Oncol Lett 2024; 27:155. [PMID: 38426158 PMCID: PMC10902755 DOI: 10.3892/ol.2024.14288] [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: 09/13/2023] [Accepted: 12/18/2023] [Indexed: 03/02/2024] Open
Abstract
The present report described the case of a 74-year-old male patient with asbestos exposure whose chest computed tomography revealed a right lower lobe nodule and right pleural effusion. Pleural biopsy led to the diagnosis of epithelial malignant pleural mesothelioma (cT2N0M0, stage IB). Combination therapy with cisplatin + pemetrexed led to the complete remission of malignant pleural mesothelioma; however, the right lower lobe nodule grew in size over time. The patient was subsequently diagnosed with lung adenocarcinoma (cT1aN0M0, stage IA1) by computed tomography-guided biopsy performed 18 months after chemotherapy initiation and achieved remission of lung adenocarcinoma with stereotactic radiotherapy. The patient was alive without recurrence at the 12-month follow-up. The present case illustrated that multiple active regimens are currently available for malignant pleural mesothelioma and lung cancer that can aid in the treatment of complex cases.
Collapse
Affiliation(s)
- Yuki Amakusa
- Department of Internal Medicine, Gamagori City Hospital, Gamagori, Aichi 443-8501, Japan
| | - Tatsuro Suzuki
- Department of Respiratory Medicine, Toyokawa City Hospital, Toyokawa, Aichi 442-8561, Japan
| | - Yu Hikosaka
- Department of Thoracic Surgery, Toyokawa City Hospital, Toyokawa, Aichi 442-8561, Japan
| | - Masaya Takemura
- Department of Internal Medicine, Gamagori City Hospital, Gamagori, Aichi 443-8501, Japan
- Department of Education and Research Center for Community Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Tetsuya Oguri
- Department of Internal Medicine, Gamagori City Hospital, Gamagori, Aichi 443-8501, Japan
- Department of Education and Research Center for Community Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| |
Collapse
|
194
|
Gamez Casado S, Benitez Fuentes JD, Álvarez Rodríguez B, García Ledo G. Achieving complete metabolic response in stage IV lung adenocarcinoma with chemotherapy, nivolumab, ipilimumab, and salvage SBRT: A case report. Respirol Case Rep 2024; 12:e01362. [PMID: 38660343 PMCID: PMC11042882 DOI: 10.1002/rcr2.1362] [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: 03/12/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024] Open
Abstract
Oncogene-negative, PDL1-negative metastatic non-small cell lung cancer (NSCLC) presents significant treatment challenges due to its complexity and resistance to conventional therapies. The case report presented addresses a 55-year-old male patient with oncogene-negative, PDL1-negative stage IV lung adenocarcinoma, showcasing an exceptional complete metabolic response to a multimodality treatment combining double immune checkpoint inhibition (ICI) and chemotherapy, followed by salvage stereotactic body radiotherapy (SBRT). The patient underwent a treatment regimen incorporating two cycles of carboplatin, pemetrexed, nivolumab, and ipilimumab followed by nivolumab, and ipilimumab maintenance. After a partial response, SBRT was applied to persistent lesions, achieving a complete metabolic response. This case highlights the potential of combining dual ICI with chemotherapy and SBRT in treating oncogene-negative, PDL1-negative NSCLC underscoring the importance of multimodality treatment strategies.
Collapse
Affiliation(s)
- Salvador Gamez Casado
- Hospital Universitario HM Sanchinarro, Centro Integral Oncológico Clara Campal (HM‐CIOCC)Department of Medical OncologyMadridSpain
| | - Javier David Benitez Fuentes
- Hospital Universitario HM Sanchinarro, Centro Integral Oncológico Clara Campal (HM‐CIOCC)Department of Medical OncologyMadridSpain
- Hospital General Universitario de ElcheDepartment of Medical OncologyElcheSpain
| | - Beatriz Álvarez Rodríguez
- Hospital Universitario HM Sanchinarro, Centro Integral Oncológico Clara Campal (HM‐CIOCC)Department of Radiation OncologyMadridSpain
| | - Gema García Ledo
- Hospital Universitario HM Sanchinarro, Centro Integral Oncológico Clara Campal (HM‐CIOCC)Department of Medical OncologyMadridSpain
| |
Collapse
|
195
|
Motta-Guerrero R, Recondo G, Cardona A, Corrales L, Arnao V, Failoc-Rojas VE, Aliaga C. The role of angiogenesis inhibitors associated with tyrosine kinase inhibitors in the first-line treatment for EGFR-mutated advanced lung cancer. Crit Rev Oncol Hematol 2024; 196:104294. [PMID: 38346461 DOI: 10.1016/j.critrevonc.2024.104294] [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: 06/15/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/29/2024] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are the standard treatment for epidermal growth factor receptor mutant (EGFRm) advanced non-small cell lung cancer (NSCLC). Combining TKIs with an angiogenesis inhibitor has shown promise in pre-clinical studies. A systematic search of clinical trials found that combining erlotinib (a first-generation TKI) with bevacizumab or ramucirumab (angiogenesis inhibitors) improved progression-free survival (PFS) in EGFRm advanced NSCLC patients compared to TKI alone. However, no significant benefit in overall survival (OS) was observed in trials. Similar efficacy was seen in patients with specific EGFR mutations. Third generation TKIs were used as second-line therapy for patients with the T790M mutation. The combination treatment was associated with a higher incidence of severe adverse events. Overall, combining erlotinib or another TKI with an angiogenesis inhibitor is a safe and effective alternative for first-line treatment in EGFRm advanced NSCLC, particularly in countries without access to osimertinib and for patients with the EGFR L858R mutation.
Collapse
Affiliation(s)
| | - Gonzalo Recondo
- Medical Oncology Department, Bradford Hill Clinical Research Center, Santiago, Chile
| | - Andres Cardona
- Direction of Research and Education / Thoracic Oncology Unit, Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center - CTIC, Bogotá, Colombia
| | - Luis Corrales
- Centro de Investigación y Manejo del Cáncer (CIMCA), San José, Costa Rica
| | - Verónica Arnao
- Instituto Nacional de Enfermedades Neoplásicas (INEN), Lima, Peru
| | | | | |
Collapse
|
196
|
Gálffy G, Morócz É, Korompay R, Hécz R, Bujdosó R, Puskás R, Lovas T, Gáspár E, Yahya K, Király P, Lohinai Z. Targeted therapeutic options in early and metastatic NSCLC-overview. Pathol Oncol Res 2024; 30:1611715. [PMID: 38605928 PMCID: PMC11006988 DOI: 10.3389/pore.2024.1611715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 02/21/2024] [Indexed: 04/13/2024]
Abstract
The complex therapeutic strategy of non-small cell lung cancer (NSCLC) has changed significantly in recent years. Disease-free survival increased significantly with immunotherapy and chemotherapy registered in perioperative treatments, as well as adjuvant registered immunotherapy and targeted therapy (osimertinib) in case of EGFR mutation. In oncogenic-addictive metastatic NSCLC, primarily in adenocarcinoma, the range of targeted therapies is expanding, with which the expected overall survival increases significantly, measured in years. By 2021, the FDA and EMA have approved targeted agents to inhibit EGFR activating mutations, T790 M resistance mutation, BRAF V600E mutation, ALK, ROS1, NTRK and RET fusion. In 2022, the range of authorized target therapies was expanded. With therapies that inhibit KRASG12C, EGFR exon 20, HER2 and MET. Until now, there was no registered targeted therapy for the KRAS mutations, which affect 30% of adenocarcinomas. Thus, the greatest expectation surrounded the inhibition of the KRAS G12C mutation, which occurs in ∼15% of NSCLC, mainly in smokers and is characterized by a poor prognosis. Sotorasib and adagrasib are approved as second-line agents after at least one prior course of chemotherapy and/or immunotherapy. Adagrasib in first-line combination with pembrolizumab immunotherapy proved more beneficial, especially in patients with high expression of PD-L1. In EGFR exon 20 insertion mutation of lung adenocarcinoma, amivantanab was registered for progression after platinum-based chemotherapy. Lung adenocarcinoma carries an EGFR exon 20, HER2 insertion mutation in 2%, for which the first targeted therapy is trastuzumab deruxtecan, in patients already treated with platinum-based chemotherapy. Two orally administered selective c-MET inhibitors, capmatinib and tepotinib, were also approved after chemotherapy in adenocarcinoma carrying MET exon 14 skipping mutations of about 3%. Incorporating reflex testing with next-generation sequencing (NGS) expands personalized therapies by identifying guideline-recommended molecular alterations.
Collapse
|
197
|
Ge Y, Zhan Y, He J, Li J, Wang J, Wei X, Wang C, Gao A, Sun Y. PD-(L)1 inhibitors plus bevacizumab and chemotherapy as first-line therapy in PD-L1-negative metastatic lung adenocarcinoma: a real-world data. J Cancer Res Clin Oncol 2024; 150:135. [PMID: 38499838 PMCID: PMC10948463 DOI: 10.1007/s00432-024-05637-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 01/30/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Chemotherapy combined with immune checkpoint inhibitors (IC), bevacizumab (BC), or both (IBC) is the preferred first-line therapy for PD-L1-negative and oncogenic-driver wild-type metastatic lung adenocarcinoma. However, the optimal strategy is still undetermined. METHODS This retrospective study enrolled PD-L1-negative metastatic lung adenocarcinoma patients from four cancer centers between January 1, 2018 and June 30, 2022. All the patients received IC, BC, or IBC as the first-line therapies. The efficacy and safety were evaluated. RESULTS A total of 205 patients were included, with 60, 83, and 62 patients in IC, BC, and IBC groups, respectively. The baseline characteristics among three groups were well balanced. Patients treated with IBC had the highest objective response rate (ORR) (43.5%) and disease control rate (DCR) (100%) relative to those treated with IC (40.4%, 84.2%) or BC (40.5%, 96.2%) (ORR: P = 0.919, DCR: P < 0.01). Compared with the IC (6.74 m) or BC (8.28 m), IBC treatment significantly improved median progression-free survival (mPFS) (9.53 m, P = 0.005). However, no difference in overall survival (OS) was observed. When stratified by different clinical and molecular information, we found that male gender, ever smoking, wild-type genes mutations, and adrenal metastasis predict superior PFS benefit when treated with IBC. In patients with liver metastasis, IBC or BC treatment displayed better PFS compared with IC. No additional adverse reactions were observed in IBC group compared with other two groups. CONCLUSION Combined IBC treatment achieved superior DCR and PFS compared with IC or BC in patients with PD-L1-negative metastatic lung adenocarcinoma, while did not increase the adverse events.
Collapse
Affiliation(s)
- Yihui Ge
- Phase I Clinical Research Center, Shandong University Cancer Center, Jinan, Shandong, China
| | - Yujing Zhan
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jie He
- Department of Oncology, People's Hospital of Zhangqiu District Jinan 250299, Shandong, People's Republic of China
| | - Juan Li
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jian Wang
- Department of Medical Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaojuan Wei
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China
| | - Chunni Wang
- Department of Thoracic Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Aiqin Gao
- Department of Thoracic Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Yuping Sun
- Phase I Clinical Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
198
|
Laguna JC, García-Pardo M, Alessi J, Barrios C, Singh N, Al-Shamsi HO, Loong H, Ferriol M, Recondo G, Mezquita L. Geographic differences in lung cancer: focus on carcinogens, genetic predisposition, and molecular epidemiology. Ther Adv Med Oncol 2024; 16:17588359241231260. [PMID: 38455708 PMCID: PMC10919138 DOI: 10.1177/17588359241231260] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/22/2024] [Indexed: 03/09/2024] Open
Abstract
Lung cancer poses a global health challenge and stands as the leading cause of cancer-related deaths worldwide. However, its incidence, mortality, and characteristics are not uniform across all regions worldwide. Understanding the factors contributing to this diversity is crucial in a prevalent disease where most cases are diagnosed in advanced stages. Hence, prevention and early diagnosis emerge as the most efficient strategies to enhance outcomes. In Western societies, tobacco consumption constitutes the primary risk factor for lung cancer, accounting for up to 90% of cases. In other geographic locations, different significant factors play a fundamental role in disease development, such as individual genetic predisposition, or exposure to other carcinogens such as radon gas, environmental pollution, occupational exposures, or specific infectious diseases. Comprehensive clinical and molecular characterization of lung cancer in recent decades has enabled us to distinguish different subtypes of lung cancer with distinct phenotypes, genotypes, immunogenicity, treatment responses, and survival rates. The ultimate goal is to prevent and individualize lung cancer management in each community and improve patient outcomes.
Collapse
Affiliation(s)
- Juan Carlos Laguna
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Miguel García-Pardo
- Department of Medical Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Joao Alessi
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute
| | - Carlos Barrios
- School of Medicine, Porto Alegre, Rio Grande do Sul, Brazil
| | - Navneet Singh
- Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | | | - Herbert Loong
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Miquel Ferriol
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
- Barcelona Neural Networking Center, Universitat Politècnica de Catalunya, Barcelona, Spain
| | | | - Laura Mezquita
- Medical Oncology Department, Hospital Clinic of Barcelona, Calle Villarroel 170, Barcelona 08036, Spain
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
199
|
Stevens S, Nindra U, Shahnam A, Wei J, Bray V, Pal A, Yip PY, Linton A, Blinman P, Nagrial A, Lee J, Boyer M, Kao S. Real world efficacy and toxicity of consolidation durvalumab following chemoradiotherapy in older Australian patients with unresectable stage III non-small cell lung cancer. J Geriatr Oncol 2024; 15:101705. [PMID: 38290173 DOI: 10.1016/j.jgo.2024.101705] [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: 07/31/2023] [Revised: 11/29/2023] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
INTRODUCTION Consolidation durvalumab following platinum-based chemoradiotherapy (CRT) significantly improved overall survival for patients with unresectable stage III non-small cell lung cancer (NSCLC) in the PACIFIC trial. However, older patients were underrepresented in PACIFIC, and subsequent analyses suggested trends toward poorer survival and increased toxicity in patients aged ≥70 years old. We assessed the effectiveness and safety of consolidation durvalumab following CRT in older Australian patients with unresectable stage III NSCLC. MATERIALS AND METHODS This retrospective observational study was conducted across seven sites in Sydney, Australia between January 2018 and September 2021. All adult patients with unresectable stage III NSCLC who received platinum-based chemoradiotherapy followed by at least one cycle of consolidation durvalumab were included. Older patients were defined as being ≥70 years old. RESULTS Of 152 patients included in the analysis, 42.8% (n = 67) patients were 70 years or older. Median follow-up was 26.1 months. The two-year overall survival and median PFS was similar between older and younger patients. At two years, 74.8% (95% confidence interval [CI]: 65.4-84.2%) of patients <70 years old and 65.2% (95% CI: 53.4-77.0%) of older patients were alive (p = 0.07; hazard ratio [HR] 1.64, 95% CI: 0.95-2.81). Median progression-free survival (PFS) in patients <70 years was 30.3 months (95% CI: 22.2-38.4 months) compared with 26.7 months (95% CI: 12.8-40.6 months) in older patients (p = 0.22; HR 1.46, 95% CI: 0.80-2.65). Toxicity was also similar, with 11.5% of patients <70 years old and 18.5% of older patients experiencing grade 3-4 adverse events (AEs; p = 0.23); 16.1% and 24.6% of the patients, respectively, discontinued treatment due to toxicity (p = 0.19). Grade 3-4 AEs and treatment discontinuation were associated with Charlson Comorbidity Index >5 (p = 0.011) and chronic obstructive pulmonary disease diagnosis at presentation (p = 0.002), respectively. DISCUSSION Older Australian patients receiving consolidation durvalumab following CRT experienced comparable outcomes to their younger peers. Comorbidity burden may be more important determinants of treatment tolerance than chronological age.
Collapse
Affiliation(s)
- Samuel Stevens
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, 119-143 Missenden Road, Camperdown, NSW 2050, Australia; Department of Medical Oncology, Concord Cancer Centre, Concord Repatriation General Hospital, Sydney, Hospital Road, Concord, NSW 2139, Australia; School of Medicine, The University of Sydney, Camperdown, NSW 2006, Australia.
| | - Udit Nindra
- Department of Medical Oncology, Liverpool Hospital, Sydney, Cnr Elizabeth and Goulburn Street, Liverpool, NSW 2170, Australia; School of Medicine, University of New South Wales, Level 2, AGSM Building, Gate 11 Botany Street, Kensington, NSW 2052, Australia
| | - Adel Shahnam
- Department of Medical Oncology, Crown Princess Margaret Cancer Centre, Westmead Hospital, Sydney, Cnr Hawkesbury and Darcy Road, Westmead, NSW, Australia, 2145
| | - Joe Wei
- Department of Medical Oncology, Crown Princess Margaret Cancer Centre, Westmead Hospital, Sydney, Cnr Hawkesbury and Darcy Road, Westmead, NSW, Australia, 2145; School of Medicine, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Victoria Bray
- Department of Medical Oncology, Liverpool Hospital, Sydney, Cnr Elizabeth and Goulburn Street, Liverpool, NSW 2170, Australia
| | - Abhijit Pal
- Department of Medical Oncology, Liverpool Hospital, Sydney, Cnr Elizabeth and Goulburn Street, Liverpool, NSW 2170, Australia; Department of Medical Oncology, Bankstown-Lidcombe Hospital, Sydney, Eldrige Road, Bankstown, NSW 2200, Australia
| | - Po Yee Yip
- Department of Medical Oncology, Macarthur Cancer Therapy Centre, Campbelltown Hospital, Sydney, Therry Road, Campbelltown, NSW 2560, Australia
| | - Anthony Linton
- Department of Medical Oncology, Concord Cancer Centre, Concord Repatriation General Hospital, Sydney, Hospital Road, Concord, NSW 2139, Australia; School of Medicine, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Prunella Blinman
- Department of Medical Oncology, Concord Cancer Centre, Concord Repatriation General Hospital, Sydney, Hospital Road, Concord, NSW 2139, Australia; School of Medicine, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Adnan Nagrial
- Department of Medical Oncology, Crown Princess Margaret Cancer Centre, Westmead Hospital, Sydney, Cnr Hawkesbury and Darcy Road, Westmead, NSW, Australia, 2145; Department of Medical Oncology, Blacktown Cancer and Haematology Centre, Blacktown Hospital, Sydney, 18 Blacktown Road, Blacktown, NSW 2148, Australia; School of Medicine, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Jenny Lee
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, 119-143 Missenden Road, Camperdown, NSW 2050, Australia; Macquarie Medical School, Macquarie University, Wallumattagal Campus, Macquarie, NSW 2109, Australia
| | - Michael Boyer
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, 119-143 Missenden Road, Camperdown, NSW 2050, Australia; School of Medicine, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Steven Kao
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, 119-143 Missenden Road, Camperdown, NSW 2050, Australia; School of Medicine, The University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
200
|
Mersiades AJ, Solomon BJ, Thomas DM, Lee CK, Cummins MM, Sebastian L, Ballinger ML, Collignon E, Turnbull OM, Yip S, Morton RL, Brown C, Wheeler PJ, Itchins M, Simes RJ, Pavlakis N. ASPiRATION: Australian observational cohort study of comprehensive genomic profiling in metastatic lung cancer tissue. Future Oncol 2024; 20:361-371. [PMID: 37767626 DOI: 10.2217/fon-2023-0366] [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] [Indexed: 09/29/2023] Open
Abstract
ASPiRATION is a national prospective observational cohort study assessing the feasibility, clinical and economic value of up-front tissue-based comprehensive genomic profiling (CGP) to identify actionable genomic alterations in participants with newly diagnosed metastatic non-squamous non-small-cell lung cancer in Australia. This study will enrol 1000 participants with tumor available for CGP and standard of care molecular testing (EGFR/ALK/ROS1). Participants with actionable variants may receive novel targeted treatments through ASPiRATION-specific substudies, other trials/programs. Clinical outcome data will be collected for a minimum of 2 years. Study outcomes are descriptive, including the ability of CGP to identify additional actionable variants, leading to personalized treatment recommendations, and will describe the feasibility, efficiency, cost and utility of implementation of CGP nationally.
Collapse
Affiliation(s)
- Antony J Mersiades
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
- Department of Medical Oncology, Northern Beaches Hospital, Frenchs Forest, NSW, 2086, Australia
| | - Benjamin J Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3001, Australia
| | - David M Thomas
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Randwick, NSW, 2031, Australia
| | - Chee K Lee
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
- Department of Medical Oncology, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Michelle M Cummins
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Lucille Sebastian
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Mandy L Ballinger
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Randwick, NSW, 2031, Australia
| | - Emily Collignon
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
| | - Olivia Mh Turnbull
- Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
| | - Sonia Yip
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Rachael L Morton
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Chris Brown
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Patrick J Wheeler
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Malinda Itchins
- Department of Medical Oncology, Royal North Shore Hospital, University of Sydney, St Leonards, NSW, 2065, Australia
| | - R John Simes
- National Health & Medical Research Council (NHMRC) Clinical Trials Centre, University of Sydney, Camperdown, NSW, 2050, Australia
| | - Nick Pavlakis
- Department of Medical Oncology, Royal North Shore Hospital, University of Sydney, St Leonards, NSW, 2065, Australia
| |
Collapse
|