1
|
He L, Li L. Case report: MSI-H, EGFR mutation, and ground-glass nodules as diffuse pulmonary hematogenous metastases. Front Immunol 2024; 15:1478205. [PMID: 39530096 PMCID: PMC11550958 DOI: 10.3389/fimmu.2024.1478205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Ground-glass nodules (GGNs) are generally considered an early stage of lung cancer. The imaging characteristics and curative efficacy of multiple GGNs as metastases remain unclear. Microsatellite instability-high (MSI-H) is a biomarker for immunotherapy. The therapeutic effect and prognosis for patients with MSI-H and Epidermal Growth Factor Receptor (EGFR)-sensitive mutation stays uncertain. Here, we report a case of a lung adenocarcinoma patient presenting with ground-glass metastases, MSI-H, and EGFR-sensitive mutation and provide clinical data on the efficacy and prognosis. We describe the predictive significance of carcinoembryonic antigen (CEA) for disease progression when there is inconsistency between treatment effectiveness and CEA changes.
Collapse
Affiliation(s)
- Liuer He
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| | - Lin Li
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College, Beijing, China
| |
Collapse
|
2
|
Sposito M, Belluomini L, Nocini R, Insolda J, Scaglione IM, Menis J, Simbolo M, Lugini A, Buzzacchino F, Verderame F, Spinnato F, Aprile G, Calvetti L, Occhipinti M, Marinelli D, Veccia A, Lombardo F, Soto Parra HJ, Ferraù F, Savastano C, Porta C, Pradelli L, Sicari E, Castellani S, Malapelle U, Novello S, Bria E, Pilotto S, Milella M. Tissue- and liquid-biopsy based NGS profiling in advanced non-small-cell lung cancer in a real-world setting: the IMMINENT study. Front Oncol 2024; 14:1436588. [PMID: 39045557 PMCID: PMC11263796 DOI: 10.3389/fonc.2024.1436588] [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: 05/22/2024] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction To date, for all non-small cell lung cancer (NSCLC) cases, it is recommended to test for driver alterations to identify actionable therapeutic targets. In this light, comprehensive genomic profiling (CGP) with next generation sequencing (NGS) has progressively gained increasing importance in clinical practice. Here, with the aim of assessing the distribution and the real-world frequency of gene alterations and their correlation with patient characteristics, we present the outcomes obtained using FoundationOne (F1CDx) and FoundationLiquid CDx (F1L/F1LCDx) NGS-based profiling in a nationwide initiative for advanced NSCLC patients. Methods F1CDx (324 genes) was used for tissue samples, and F1L (70 genes) or F1LCDx (324 genes) for liquid biopsy, aiming to explore the real-world occurrence of molecular alterations in aNSCLC and their relationship with patients' characteristics. Results Overall, 232 advanced NSCLC patients from 11 Institutions were gathered [median age 63 years; never/former or current smokers 29.3/65.9%; adenocarcinoma/squamous 79.3/12.5%; F1CDx/F1L+F1LCDx 59.5/40.5%]. Alterations were found in 170 different genes. Median number of mutated genes per sample was 4 (IQR 3-6) and 2 (IQR 1-3) in the F1CDx and F1L/F1LCDx cohorts, respectively. TP53 (58%), KRAS (22%), CDKN2A/B (19%), and STK11 (17%) alterations were the most frequently detected. Actionability rates (tier I and II) were comparable: 36.2% F1CDx vs. 34% ctDNA NGS assays (29.5% and 40.9% F1L and F1LCDx, respectively). Alterations in KEAP1 were significantly associated with STK11 and KRAS, so as TP53 with RB1. Median tumor mutational burden was 6 (IQR 3-10) and was significantly higher in smokers. Median OS from metastatic diagnosis was 23 months (IQR 18.5-19.5) and significantly lower in patients harboring ≥3 gene mutations. Conditional three-year survival probabilities increased over time for patients profiled at initial diagnosis and exceeded those of individuals tested later in their clinical history after 12 months. Conclusion This study confirms that NGS-based molecular profiling of aNSCLC on tissue or blood samples offers valuable predictive and prognostic insights.
Collapse
Affiliation(s)
- Marco Sposito
- Section of Innovation Biomedicine – Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust/Azienda Ospedaliero-Universitaria Integrata (AOUI), Verona, Italy
| | - Lorenzo Belluomini
- Section of Innovation Biomedicine – Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust/Azienda Ospedaliero-Universitaria Integrata (AOUI), Verona, Italy
| | - Riccardo Nocini
- Otolaryngology-Head and Neck Surgery Department, University of Verona Hospital Trust, Verona, Italy
| | - Jessica Insolda
- Section of Innovation Biomedicine – Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust/Azienda Ospedaliero-Universitaria Integrata (AOUI), Verona, Italy
| | - Ilaria Mariangela Scaglione
- Section of Innovation Biomedicine – Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust/Azienda Ospedaliero-Universitaria Integrata (AOUI), Verona, Italy
| | - Jessica Menis
- Section of Innovation Biomedicine – Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust/Azienda Ospedaliero-Universitaria Integrata (AOUI), Verona, Italy
| | - Michele Simbolo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Antonio Lugini
- Medical Oncology Unit, Azienda Ospedaliera (AO) San Giovanni Addolorata Hospital, Rome, Italy
| | | | - Francesco Verderame
- Section of Oncology, Azienda Ospedaliera (AO) Ospedali Riuniti “Villa Sofia- V. Cervello”, Palermo, Italy
| | - Francesca Spinnato
- Section of Oncology, Azienda Ospedaliera (AO) Ospedali Riuniti “Villa Sofia- V. Cervello”, Palermo, Italy
| | - Giuseppe Aprile
- Department of Clinical Oncology, San Bortolo General Hospital, Azienda ULSS8 Berica, Vicenza, Italy
| | - Lorenzo Calvetti
- Department of Clinical Oncology, San Bortolo General Hospital, Azienda ULSS8 Berica, Vicenza, Italy
| | - Mario Occhipinti
- Department of Experimental Medicine, Sapienza University, Rome, Italy
- Medical Oncology Department, Fondazione Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale Dei Tumori, Milan, Italy
| | - Daniele Marinelli
- Department of Experimental Medicine, Sapienza University, Rome, Italy
- Division of Medical Oncology B, Policlinico Umberto I, Rome, Italy
| | - Antonello Veccia
- Medical Oncology Department, Santa Chiara Hospital, Trento, Italy
| | | | - Hector José Soto Parra
- Medical Oncology, Azienda Ospedaliero Universitaria Policlinico “G. Rodolico-S. Marco”, Catania, Italy
| | - Francesco Ferraù
- Department of Medical Oncology, Unità Operativa Complessa (UOC) Oncologia, Taormina, Italy
| | | | - Camilla Porta
- AdRes Health Economics and Outcome Research, Turin, Italy
| | | | | | | | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Silvia Novello
- Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Roma, Italy
- Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Sara Pilotto
- Section of Innovation Biomedicine – Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust/Azienda Ospedaliero-Universitaria Integrata (AOUI), Verona, Italy
| | - Michele Milella
- Section of Innovation Biomedicine – Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and University and Hospital Trust/Azienda Ospedaliero-Universitaria Integrata (AOUI), Verona, Italy
| |
Collapse
|
3
|
Rosca OC, Vele OE. Microsatellite Instability, Mismatch Repair, and Tumor Mutation Burden in Lung Cancer. Surg Pathol Clin 2024; 17:295-305. [PMID: 38692812 DOI: 10.1016/j.path.2023.11.011] [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] [Indexed: 05/03/2024]
Abstract
Since US Food and Drug Administration approval of programmed death ligand 1 (PD-L1) as the first companion diagnostic for immune checkpoint inhibitors (ICIs) in non-small cell lung cancer, many patients have experienced increased overall survival. To improve selection of ICI responders versus nonresponders, microsatellite instability/mismatch repair deficiency (MSI/MMR) and tumor mutation burden (TMB) came into play. Clinical data show PD-L1, MSI/MMR, and TMB are independent predictive immunotherapy biomarkers. Harmonization of testing methodologies, optimization of assay design, and results analysis are ongoing. Future algorithms to determine immunotherapy eligibility might involve complementary use of current and novel biomarkers. Artificial intelligence could facilitate algorithm implementation to convert complex genetic data into recommendations for specific ICIs.
Collapse
Affiliation(s)
- Oana C Rosca
- Molecular Pathologist/Cytopathologist, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell; Department of Pathology and Laboratory Medicine, 2200 Northern Boulevard, Suite 104, Greenvale, NY 11548, USA.
| | - Oana E Vele
- Molecular Pathologist/Cytopathologist, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell; Department of Pathology and Laboratory Medicine, Lenox Hill Hospital, New York, NY 10075, USA
| |
Collapse
|
4
|
Olgun P, Diker O. Sixth-Week Immune-Nutritional-Inflammatory Biomarkers: Can They Predict Clinical Outcomes in Patients with Advanced Non-Small Cell Lung Cancer Treated with Immune Checkpoint Inhibitors? Curr Oncol 2023; 30:10539-10549. [PMID: 38132403 PMCID: PMC10742278 DOI: 10.3390/curroncol30120769] [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: 11/05/2023] [Revised: 12/10/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND We investigated the relationships between inflammatory markers such as the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), Lung Immune Prognostic Index (LIPI), and modified Glasgow prognostic score (mGPS) to determine whether they could predict treatment response to pembrolizumab or nivolumab (immunotherapy) 6 weeks after the start of treatment (post-treatment). METHODS We included all patients with lung cancer treated with immunotherapy. We examined the biomarker trends and explored their associations with progression-free survival (PFS), overall survival (OS), and response rate (RR) at 6 weeks. RESULTS Eighty-three patients were enrolled in the study. The presence of liver metastasis, low post-treatment NLR (<5), low post-treatment PLR (<170), intermediate post-treatment LIPI, and immune-related adverse events were significantly associated with the response. The multivariate analysis revealed that high post-treatment NLRs ≥ 5 (p = 0.004) and PLRs ≥ 170 (p ≤ 0.001) were independent prognostic factors of shorter OS. A good LIPI status was associated with better PFS (p = 0.020) and OS (p = 0.065). Post-treatment mGPS (0-2) was significantly associated with improved PFS (p = 0.009) and OS (p = 0.064). CONCLUSIONS Post-treatment NLR, PLR, LIPI, and mGPS are associated with worse OS and recurrence. These findings should be independently and prospectively validated in further studies.
Collapse
Affiliation(s)
- Polat Olgun
- Department of Medical Oncology, Faculty of Medicine, Near East University, 99138 Nicosia, Cyprus;
- Medical Oncology, Dr. Burhan Nalbantoğlu Government Hospital, 99010 Nicosia, Cyprus
| | - Omer Diker
- Department of Medical Oncology, Faculty of Medicine, Near East University, 99138 Nicosia, Cyprus;
- Medical Oncology, Dr. Burhan Nalbantoğlu Government Hospital, 99010 Nicosia, Cyprus
| |
Collapse
|