1
|
Chouaid C, Grossi F, Ta Thanh Minh C, Raymond R, Bosch-Barrera J. Pooled analysis of oral vinorelbine as single agents in patients with advanced NSCLC. Lung Cancer Manag 2025; 14:2477418. [PMID: 40116568 PMCID: PMC11938966 DOI: 10.1080/17581966.2025.2477418] [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: 06/21/2024] [Accepted: 03/06/2025] [Indexed: 03/23/2025] Open
Abstract
OBJECTIVES This was a pooled analysis of data from weekly vinorelbine (VNR) treatment arms of four individual open-label, phase II studies to assess and refine the efficacy and tolerance of weekly oral VNR in a larger cohort of patients with advanced NSCLC. MATERIALS AND METHODS All patients included in this pooled analysis received oral VNR at the dose of 60 mg/m2 weekly at cycle 1 (3-week cycle), followed by an increase to 80 mg/m2 weekly for subsequent cycles until disease progression or toxicity. Efficacy was based on objective response rate (ORR), progression-free survival (PFS), and disease control rate (DCR). RESULTS A total of 247 patients were included. The ORR and DCR were 8.9% and 57.5% respectively, median PFS and OS were 3.3 and 8.5 months, respectively. Less than half (40.7%) of patients reported ≥1 serious AE (regardless of causality), with 12.3% reporting ≥1 treatment-related serious AE (grade ≥3: 11.1%). The most reported grade ≥3 AEs were neutropenia (17.6%), fatigue (5.8%), and decreased appetite (4.9%). CONCLUSION This pooled analysis showed that weekly oral VRN is a valid option, with an acceptable safety profile, in this population of patients with advanced NSCLC, confirming results from previous individual studies.
Collapse
Affiliation(s)
| | - Francesco Grossi
- Department of Medicine and Technological Innovation, Università degli Studi dell’Insubria, Varese - Medical Oncology Division, ASST Sette Laghi, Varese, Italy
| | | | - Romain Raymond
- Medical & Patient/Consumer Division, Pierre Fabre, Boulogne-Billancourt, France
| | - 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
| |
Collapse
|
2
|
Attili I, Corvaja C, Trillo Aliaga P, Del Signore E, Spitaleri G, Passaro A, de Marinis F. Dealing with KRAS G12C inhibition in non-small cell lung cancer (NSCLC) - biology, clinical results and future directions. Cancer Treat Rev 2025; 137:102957. [PMID: 40381528 DOI: 10.1016/j.ctrv.2025.102957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
KRAS G12C mutation occurs in ∼ 14 % of non-small cell lung cancer (NSCLC) patients and has been historically deemed undruggable, with immune-checkpoint inhibitors (ICIs) and platinum-based chemotherapy (PBC) representing the standard-of-care in the advanced setting. First-in-class, covalent KRAS G12C OFF-inhibitors sotorasib and adagrasib have revolutionized the therapeutic landscape and recently entered clinical practice. However, limited efficacy alongside toxicity profiles strengthen the need to design novel molecules and to optimize therapeutic strategies to address and overcome intrinsic and acquired resistance mechanisms. Moreover, KRAS G12C frequently co-occurs with STK11/KEAP1 mutations, that represent a negative prognostic factor, being associated with increased metastatic potential and reduced overall survival and poorer outcomes with ICIs. Furthermore, the high incidence of brain metastases is common in KRAS G12C-mutant NSCLC, and the efficacy of standard therapies and KRAS G12C inhibitors in treating or preventing central nervous system involvement is still suboptimal. In this context, novel inhibitors, such as broad-spectrum inhibitors targeting the active GTP-bound ON-state, pan-RAS ON inhibitors and allele-selective tricomplex inhibitors, have showed promising early clinical activity although their clinical utility needs to be further elucidated. In addition, targeting upstream, downstream and parallel signaling pathways through combination strategies might enhance the activity of KRAS G12C inhibitors and eventually improve clinical outcomes in this subset of NSCLC patients. Several combinations are currently under clinical investigation and promising approaches include combinations of KRAS G12C inhibitors with ICIs, SOS1, SHP2 inhibitors and PBC. Notwithstanding the potential improved efficacy of combination strategies, tolerability remains a critical challenge that must be carefully assessed and managed.
Collapse
Affiliation(s)
- Ilaria Attili
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Carla Corvaja
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Pamela Trillo Aliaga
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Ester Del Signore
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Gianluca Spitaleri
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy.
| | - Filippo de Marinis
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| |
Collapse
|
3
|
Suay G, Martín-Martorell P, Aparisi F, Arnal M, Guirado M, Azkárate A, Garde-Noguera J, Cumplido-Burón JD, Insa A, González-Muñoz JF, Palanca S, Díaz M, Sánchez-Hernández A, Juan-Vidal Ó. A real‑world study of clinical characteristics, treatment sequence and outcomes of patients with non-small cell lung cancer and EGFR exon 20 insertion mutations. Clin Transl Oncol 2025; 27:2568-2578. [PMID: 39499485 DOI: 10.1007/s12094-024-03776-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/18/2024] [Indexed: 11/07/2024]
Abstract
OBJECTIVES EGFR exon 20 insertion (EGFRex20ins) mutations are found in up to 4% of all patients with non-small cell lung cancer (NSCLC). These patients are often insensitive to EGFR-tyrosine kinase inhibitors (TKIs) and have worse prognosis than patients with more common EGFR mutations. In this multicenter, retrospective, real-world study, we sought to determine whether the administration of recently approved treatments that specifically target EGFRex20ins mutations could significantly improve outcomes in this patient population. MATERIALS AND METHODS We evaluated the clinical features of 41 patients diagnosed with NSCLC and EGFRex20ins mutations, their evolution, and response to treatments received across 7 hospitals in the Valencian Community, Spain, between 31st December 2012 and 31st December 2022. RESULTS 32 patients (72%) developed metastatic disease, and 29 (71%) of them received oncological treatment. We found that administering a targeted therapy against EGFRex20ins mutations (amivantamab, mobocertinib and/or sunvozertinib) at some point during the course of treatment, significantly increased the median OS of metastatic patients from 8 months (95% CI 0-21.7) to 30 months (95% CI 11.1-48.8; Hazard ratio = 0.297, p = 0.02). CONCLUSION Our findings contribute to the evolving standard of care for this specific population and highlight the clinical benefits of targeted cancer therapies.
Collapse
Affiliation(s)
- Guillermo Suay
- Medical Oncology Department, Biomarker and Precision Medicine Unit, La Fe University and Polytechnic Hospital, Valencia, Spain
| | | | - Francisco Aparisi
- Medical Oncology Investigator, Biomarker and Precision Medicine Unit, Health Research Institute Hospital La Fe, Valencia, Spain
| | - María Arnal
- Medical Oncology Department, Provincial Hospital Consortium, Castellón, Spain
| | - María Guirado
- Medical Oncology Department, General University Hospital of Elche, Alicante, Spain
| | - Aitor Azkárate
- Medical Oncology Department, Son Espases University Hospital, Palma, Spain
| | | | | | - Amelia Insa
- Medical Oncology Department, University Clinical Hospital, Valencia, Spain
| | | | - Sarai Palanca
- Clinical Analysis Department, Molecular Biology Unit, La Fe University and Polytechnic Hospital, Valencia, Spain
| | - María Díaz
- Clinical Analysis Department, Castellón University General Hospital, Castellón de La Plana, Spain
| | | | - Óscar Juan-Vidal
- Medical Oncology Department, Biomarker and Precision Medicine Unit, La Fe University and Polytechnic Hospital, Valencia, Spain.
| |
Collapse
|
4
|
Borea R, Reduzzi C. The growing field of liquid biopsy and its Snowball effect on reshaping cancer management. THE JOURNAL OF LIQUID BIOPSY 2025; 8:100293. [PMID: 40255897 PMCID: PMC12008596 DOI: 10.1016/j.jlb.2025.100293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/22/2025]
Abstract
Liquid biopsy (LB) has emerged as a transformative tool in oncology, providing a minimally invasive approach for tumor detection, molecular characterization, and real-time treatment monitoring. By analyzing circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), extracellular vesicles (EVs), and microRNA (miRNA), LB enables comprehensive tumor profiling without the need for traditional tissue biopsies. Over the past decade, research in this field has expanded exponentially, leading to the integration of LB into clinical practice for specific cancer types, including lung and breast cancer. In 2024, the Journal of Liquid Biopsy (JLB) published innovative studies exploring the latest advancements in LB technologies, biomarkers, and their applications for cancer detection, minimal residual disease (MRD) monitoring, and therapy response assessment. This review synthesizes recent findings on the role of LB in cancer treatment and monitoring across different biomarkers, with a particular focus on newly published studies and their context within translational research. Additionally, it highlights emerging techniques such as fragmentomics, artificial intelligence, and multiomics, paving the way for more precise, personalized treatment decisions. Despite these advancements, challenges remain in standardizing methodologies, optimizing clinical validation, and integrating LB into routine oncological workflows. This mini-review highlights the evolving landscape of LB research and its potential to revolutionize cancer diagnosis, treatment monitoring, and therapeutic decision-making, ushering in a new era of precision oncology.
Collapse
Affiliation(s)
- Roberto Borea
- Department of Public Health, University Federico II of Naples, Naples, Italy
- Department of Internal Medicine and Medical Sciences (DiMI), School of Medicine, University of Genova, Genova, Italy
| | - Carolina Reduzzi
- Department of Medicine, Weill Cornell Medicine, Englander Institute for Precision Medicine, New York Presbyterian Hospital, New York, NY, 10021, USA
| |
Collapse
|
5
|
Guidi L, Etessami J, Valenza C, Valdivia A, Meric-Bernstam F, Felip E, Curigliano G. Bispecific Antibodies in Hematologic and Solid Tumors: Current Landscape and Therapeutic Advances. Am Soc Clin Oncol Educ Book 2025; 45:e473148. [PMID: 40198874 DOI: 10.1200/edbk-25-473148] [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: 04/10/2025]
Abstract
Bispecific antibodies (bsAbs) have emerged as a novel class of therapeutics, offering a dual-targeting strategy to enhance the therapeutic efficacy of monoclonal antibodies, which is often limited by tumor heterogeneity and the occurrence of resistance mechanisms. By simultaneously engaging two distinct antigens or pathways, bsAbs disrupt multiple signaling cascades simultaneously, preventing escape mechanisms and offering a more durable response. Furthermore, they can optimize immune activation, improving immune cell recruitment strategies. In particular, T-cell engager bsAbs facilitate immune cell-mediated tumor destruction by linking T cells to tumor antigens. Instead, dual immune checkpoint inhibitors (CPIs) enhance immune activation by blocking inhibitory signals. Additionally, bsAbs targeting tumor growth factors or receptor tyrosine kinases offer solutions for overcoming drug resistance in solid tumors. Although bsAbs have shown remarkable success in hematologic malignancies, their expansion into solid tumors faces key challenges, including tumor heterogeneity, limited tumor penetration, and the risk of on-target, off-tumor toxicities. Addressing these challenges requires innovative engineering strategies, optimized delivery mechanisms, and careful patient selection to maximize therapeutic benefit while mitigating adverse effects. The efficacy of bsAbs in clinical trials has led to their approval for both hematologic and solid malignancies, with numerous agents in development. Combination strategies with chemotherapy, targeted agents, and immune CPIs could represent a promising strategy to further expand their potential. As research progresses, bsAbs are expected to play a role in reshaping the future of precision oncology, offering more effective and tailored treatment options.
Collapse
Affiliation(s)
- Lorenzo Guidi
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Julian Etessami
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA
| | - Augusto Valdivia
- Department of Medical Oncology, Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Enriqueta Felip
- Department of Medical Oncology, Vall d'Hebron Hospital Campus and Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
6
|
Brandt WS. Genomic Alterations of Early-Onset Lung Adenocarcinoma: A Step in the Right Direction. Ann Thorac Surg 2025; 119:1204-1205. [PMID: 39515737 DOI: 10.1016/j.athoracsur.2024.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Affiliation(s)
- Whitney S Brandt
- Washington University St Louis, 660 Euclid Ave, St Louis, MO 63110.
| |
Collapse
|
7
|
Trisolini R, Cetoretta V, Sotgiu G, Cancellieri A, Puci M, Viscuso M, Livi V, Cani M, Scambia G, Cappuzzo F, Bria E, Novello S. Supraclavicular Lymph Node Metastases in Advanced Lung Cancer: Prevalence and Analysis of Demographic, Clinical and Molecular Characteristics. Clin Lung Cancer 2025; 26:e284-e292. [PMID: 40037966 DOI: 10.1016/j.cllc.2025.02.003] [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: 08/29/2024] [Revised: 01/29/2025] [Accepted: 02/07/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND The prevalence of supraclavicular lymph nodes metastases (SNM) in advanced lung cancer has not been systematically evaluated, nor has then been a comparison of demographic, clinical, or molecular characteristics between patients with and without SNM. METHODS In this prospective cohort study, the presence of SNM was evaluated using imaging studies (CT, PET, neck ultrasonography) in patients with suspected advanced lung cancer referred for biopsy aimed at diagnosis and molecular profiling. Ultrasound-guided biopsy confirmed or excluded metastatic involvement when suspicious supraclavicular nodes were identified. We assessed the prevalence of SNM and compared the demographic, clinicopathologic and molecular characteristics of patients with and without SNM. RESULTS Among the 348 patients with advanced lung cancer, 94 (27%) had SMN. SMN was more common in small cell lung cancer (24/48, 50%) and adenocarcinoma (61/248, 24.6%) than in squamous cell carcinoma (4/35, 11.4%). Compared to patients without SMN, those with SMN were more likely to have small-cell lung cancer, N2/3 disease (97.9 vs. 83.9%, P < .0001), liver metastases (29.8% vs. 16.1% P = .006), and metastases to less common sites (33.7% vs. 14.1%, P < .0001). The prevalence of genomic alterations and PD-L1 expression did not differ between biopsy samples obtained from SNM and those from the primary tumor or other metastatic sites. CONCLUSION SNM is common in patients with advanced small-cell lung cancer and adenocarcinoma. Ultrasound-guided biopsy of SNM is a simple and relatively inexpensive method for obtaining adequate tissue samples for diagnosis and comprehensive molecular profiling.
Collapse
Affiliation(s)
- Rocco Trisolini
- Interventional Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Hearth, Rome, Italy
| | - Valeria Cetoretta
- Interventional Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Hearth, Rome, Italy; Department of Oncology, University of Turin, Azienda Ospedaliero-Universitaria San Luigi, Turin, Italy
| | - Giovanni Sotgiu
- Clinical Epidemiology and Medical Statistics Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | | | - Mariangela Puci
- Clinical Epidemiology and Medical Statistics Unit, Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Marta Viscuso
- Interventional Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Hearth, Rome, Italy; Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Vanina Livi
- Interventional Pulmonology Division, Fondazione Policlinico Universitario A. Gemelli IRCCS, Catholic University of the Sacred Hearth, Rome, Italy
| | - Massimiliano Cani
- Department of Oncology, University of Turin, Azienda Ospedaliero-Universitaria San Luigi, Turin, Italy
| | - Giovanni Scambia
- Gynecologic Oncology Unit, Department of Woman, Child and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Federico Cappuzzo
- Medical Oncology Division, IRCCS Regina Elena National Cancer Institute, Rome, Italy Medical
| | - Emilio Bria
- Oncology Division, Gemelli Isola Hospital, Catholic University of the Sacred Hearth, Rome, Italy
| | - Silvia Novello
- Department of Oncology, University of Turin, Azienda Ospedaliero-Universitaria San Luigi, Turin, Italy.
| |
Collapse
|
8
|
Bestvina CM, Kim C, Daaboul N. Driving Best Practices Throughout the Treatment Journey for Patients with NSCLC with Actionable Alterations: A Podcast Discussion. Adv Ther 2025:10.1007/s12325-025-03195-7. [PMID: 40402375 DOI: 10.1007/s12325-025-03195-7] [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: 10/10/2024] [Accepted: 04/01/2025] [Indexed: 05/23/2025]
Abstract
Non-small cell lung cancer (NSCLC) treatment has been revolutionized by the advent of targeted therapies for tumors harboring specific actionable alterations. Targeted agents are now approved for use in patients with advanced NSCLC with various drivers including ALK rearrangements, BRAF V600E mutations, EGFR mutations, ERBB2 mutations, KRAS G12C mutations, MET exon 14 skipping alterations, NTRK fusions, RET rearrangements, and ROS1 rearrangements. Importantly, the availability of these agents has raised the clinical question of how to optimally sequence their use alongside chemotherapy and/or immunotherapy strategies, which are indicated for broader populations. Key considerations include (i) evidence for better outcomes when first-line treatment is initiated following availability of molecular profiling data; (ii) the decreasing proportion of patients able to receive therapy in each successive treatment line; (iii) the efficacy of targeted agents demonstrated in either single-arm trials or head-to-head comparisons with chemotherapy and/or immunotherapy, as compared with evidence for poor or modest efficacy of immunotherapy in patients with tumors with actionable drivers; (iv) real-world data showing better outcomes of patients with tumors with actionable alterations who received targeted therapies compared with those who did not; (v) the generally favorable safety profile of targeted therapies, as well as the potential for increased toxicity when immunotherapy precedes certain targeted agents; and (vi) patient-centric factors including the greater ease of administration of oral targeted therapies over intravenous chemotherapy or immunotherapy strategies. In line with these considerations, guidelines typically recommend most targeted agents approved for first-line use as initial therapy over chemotherapy and/or immunotherapy. In this podcast, the authors discuss the current therapeutic landscape of NSCLC with actionable alterations and provide their perspectives on treatment algorithms, and how to optimally sequence therapies for patients with tumors harboring actionable alterations, using patient cases to illustrate key principles.
Collapse
Affiliation(s)
- Christine M Bestvina
- Department of Medicine, University of Chicago Medical Center, 5841 S Maryland Ave, MC 2115, Chicago, IL, 60637, USA.
| | - Chul Kim
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Nathalie Daaboul
- Centre Intégré de Cancérologie de la Montérégie, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
9
|
Gautschi O, Park K, Solomon BJ, Tomasini P, Loong HH, De Braud F, Goto K, Peterson P, Barker S, Liming K, Oxnard GR, Frimodt-Moller B, Drilon A. Selpercatinib in RET Fusion-Positive Non-Small Cell Lung Cancer: Final Safety and Efficacy, Including Overall Survival, From the LIBRETTO-001 Phase I/II Trial. J Clin Oncol 2025; 43:1758-1764. [PMID: 39983053 PMCID: PMC12084017 DOI: 10.1200/jco-24-02076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/19/2024] [Accepted: 01/15/2025] [Indexed: 02/23/2025] Open
Abstract
LIBRETTO-001 (ClinicalTrials.gov identifier: NCT03157128) is a registrational phase I/II, single-arm, open-label trial of selpercatinib in RET-dependent cancers. With 19 months of additional follow-up, we report the final efficacy and safety results of selpercatinib in patients with RET fusion-positive non-small cell lung cancer (NSCLC) who had previously received platinum-based chemotherapy (N = 247) or were treatment-naïve (N = 69). The objective response rate (ORR) was 62% for pretreated patients and 83% for treatment-naïve patients. Duration of response (DoR) was 31.6 months for pretreated and 20.3 months for treatment-naïve patients (median follow-up approximately 38 months). Median progression-free survival (PFS) was 26.2 months for pretreated and 22.0 months for treatment-naïve patients (median follow-up approximately 40 months). Median overall survival was 47.6 months in pretreated patients and was not reached in the treatment-naïve group (median follow-up approximately 43 months). At the 3-year landmark estimate, 57% of pretreated and 66% of treatment-naïve patients were alive. Among 26 patients with measurable CNS metastases at baseline, the CNS-ORR was 85% with a CNS-DoR of 9.4 months and CNS-PFS of 11.0 months. The safety profile of selpercatinib was consistent with previous reports. With substantial additional follow-up, selpercatinib continued to show durable responses and intracranial activity, with a manageable safety profile in patients with RET fusion-positive NSCLC.
Collapse
Affiliation(s)
- Oliver Gautschi
- University of Berne and Medical Oncology, Cantonal Hospital of Lucerne, Lucerne, Switzerland
| | - Keunchil Park
- Division of Heamatology/Oncology, Medicine, Samsung Medical Center (SMC)—Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Thoracic/Head & Neck Medical Oncology, UT MD Anderson Cancer Center, Houston, TX
| | | | | | - Herbert H. Loong
- Department of Clinical Oncology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Filippo De Braud
- Medical Oncology & Haematology Department, Fondazione IRCCS—Istituto Nazionale dei Tumori, Milan, Italy
| | - Koichi Goto
- National Cancer Center Hospital East, Kashiwa, Japan
| | | | | | | | | | | | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical Center, New York, NY
| |
Collapse
|
10
|
Lee JH, Seo SH, Shim J, Kim YN, Yoon K. Narciclasine enhances cisplatin-induced apoptotic cell death by inducing unfolded protein response-mediated regulation of NOXA and MCL1. Cell Mol Biol Lett 2025; 30:59. [PMID: 40369444 PMCID: PMC12076939 DOI: 10.1186/s11658-025-00735-5] [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: 10/24/2024] [Accepted: 04/22/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Platinum-based chemotherapy is commonly used to treat non-small cell lung cancer (NSCLC); however, innate and acquired resistance is clinically seen in many patients. Hence, a combinatorial approach with novel therapeutic agents to overcome chemoresistance is a promising option for improving patient outcomes. We investigated the combinational anticancer efficacy of cisplatin and narciclasine in three-dimensional NSCLC tumor spheroids. METHODS To assess the efficacy of cisplatin and narciclasine, cell viability assays, live/dead cell staining, cell death enzyme-linked immunosorbent assay (ELISA), western blot analysis for proteins related to apoptosis, and in vivo xenograft experiments were performed. The synergistic effects of cisplatin and narciclasine were elucidated through transcriptomic analysis and subsequent validation of candidate molecules by regulating their expression. To clarify the underlying molecular mechanisms, the activation of unfolded protein responses and kinetics of a candidate protein were assessed. RESULTS Narciclasine inhibited viability of NSCLC tumor spheroids and augmented the sensitivity of cisplatin-resistant tumor spheroids to cisplatin by inducing apoptosis. After conducting bioinformatic analysis using RNA sequencing data and functional validation experiments, we identified NOXA as a key gene responsible for the enhanced apoptosis observed with the combination of cisplatin and narciclasine. This treatment dramatically increased NOXA while downregulating anti-apoptotic MCL1 levels. Silencing NOXA reversed the enhanced apoptosis and restored MCL1 levels, while MCL1 overexpression protected tumor spheroids from combination treatment-induced apoptosis. Interestingly, narciclasine alone and in combination with cisplatin induced unfolded protein response and inhibited general protein synthesis. Furthermore, the combination treatment increased NOXA expression through the IRE1α-JNK/p38 axis and the activation of p53. Cisplatin alone and in combination with narciclasine destabilized MCL1 via NOXA-mediated proteasomal degradation. CONCLUSIONS We identified a natural product, narciclasine, that synergizes with cisplatin. The combination of cisplatin and narciclasine induced NOXA expression, downregulated MCL1, and ultimately induced apoptosis in NSCLC tumor spheroids. Our findings suggest that narciclasine is a potential natural product for combination with cisplatin for treatment of NSCLC.
Collapse
Affiliation(s)
- Ji Hae Lee
- Cancer Metastasis Branch, Research Institute, National Cancer Center, Goyang, 10408, South Korea
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, South Korea
| | - Seung Hee Seo
- Cancer Metastasis Branch, Research Institute, National Cancer Center, Goyang, 10408, South Korea
| | - Jaegal Shim
- Cancer Metastasis Branch, Research Institute, National Cancer Center, Goyang, 10408, South Korea
| | - Yong-Nyun Kim
- Cancer Metastasis Branch, Research Institute, National Cancer Center, Goyang, 10408, South Korea
| | - Kyungsil Yoon
- Cancer Metastasis Branch, Research Institute, National Cancer Center, Goyang, 10408, South Korea.
| |
Collapse
|
11
|
Pan Y, Yan L, Wang S, Li H, Chen Q. Case Report: Ensartinib as a first-line treatment for SMARCA4-deficient and EML4-ALK non-small cell lung cancer. Front Oncol 2025; 15:1530142. [PMID: 40416876 PMCID: PMC12098044 DOI: 10.3389/fonc.2025.1530142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 04/14/2025] [Indexed: 05/27/2025] Open
Abstract
SMARC4 is the catalytic subunit of the SWI/SNF chromatin remodeling complex and is one of the most common altered chromatin remodeling ATPases in cancer. Studies have indicated that SMARCA4 loss is associated with highly aggressive tumors, independently predicting shorter overall and disease-specific survival. SMARCA4-deficient non-small cell lung cancer (NSCLC) primarily affects male individuals, especially smokers, and is characterized by large, aggressive tumors. Cases of SMARCA4 deletion combined with actionable driver gene mutations (e.g., ALK) are rarely reported. In this report, we describe a male non-smoker diagnosed with SMARCA4-deficient, EML4-ALK non-small cell lung cancer who has been undergoing ensartinib targeted therapy for 3 months, resulting in a significant partial response. We also propose that, from a signaling perspective, the presence of SMARCA4 deficiency may influence the sensitivity of EML4-ALK NSCLC to targeted therapy, highlighting the need for further investigation into the underlying mechanisms and the exploration of novel therapeutic approaches.
Collapse
Affiliation(s)
- Yanqing Pan
- Department of Respiratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Lingxin Yan
- Department of Respiratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shaoxi Wang
- Department of Respiratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Huiling Li
- Department of Respiratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Quanfang Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
12
|
Damuzzo V, Gasperoni L, Del Bono L, Ossato A, Inno A, Messori A. Treatment of metastatic ALK-positive non-small cell lung cancer: indirect comparison of different ALK inhibitors using reconstructed patient data. Front Oncol 2025; 15:1566816. [PMID: 40416880 PMCID: PMC12098575 DOI: 10.3389/fonc.2025.1566816] [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: 01/25/2025] [Accepted: 04/14/2025] [Indexed: 05/27/2025] Open
Abstract
Introduction Anaplastic lymphoma kinase (ALK) inhibitors (ALKi) are the standard treatment for metastatic, ALK-positive non-small cell lung cancer (NSCLC). Second- and third-generation ALKi, including alectinib, brigatinib, ensartinib, envonalkib, and lorlatinib, have shown better efficacy than crizotinib. However, due to the lack of direct head-to-head comparisons among these agents, the optimal treatment for metastatic ALK-positive NSCLC remains unclear. Methods This study used the IPDfromKM (Individual Patient Data from Kaplan-Meier) method to reconstruct patient-level data from Kaplan-Meier curves of seven randomized phase III trials, involving a total of 3,850 patients. Crizotinib arms were pooled as the common comparator. Progression-free survival (PFS) was the primary endpoint, assessed using Cox proportional hazards models and restricted mean survival time (RMST). Subgroup analyses focused on patients with baseline central nervous system (CNS) metastases. Results All ALKi significantly improved PFS compared to crizotinib. Lorlatinib showed the most meaningful improvement, with the greatest benefit in both overall PFS (HR=0.28; 95% CI 0.21-0.38) and CNS PFS (HR=0.09; 95% CI 0.04-0.2). In direct comparisons, lorlatinib outperformed brigatinib (HR=0.59; 95% CI 0.39-0.87) and envonalkib (HR=0.52; 95% CI 0.35-0.77) in terms of PFS. While lorlatinib also showed improved PFS compared to alectinib (HR=0.72; 95% CI 0.50-1.04) and ensartinib (HR=0.73; 95% CI 0.48-1.10), these differences were not statistically significant. Lorlatinib demonstrated the greatest benefit in PFS among patients with baseline CNS metastases. Conclusion In this indirect comparison using reconstructed patient data, lorlatinib emerged as the most effective ALKi, showed the most favorable HR for PFS compared to the other ALKi, although it did not reach statistical significance versus alectinib and ensartinib. Additionally, lorlatinib showed the highest efficacy in the control of CNS progression.
Collapse
Affiliation(s)
- Vera Damuzzo
- Hospital Pharmacy, Vittorio Veneto Hospital, AULSS2 Marca Trevigiana, Vittorio Veneto, Italy
- Italian Society of Clinical Pharmacy and Therapeutics (SIFaCT), Turin, Italy
| | - Lorenzo Gasperoni
- Oncological Pharmacy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Luna Del Bono
- Department of Pharmacy, School of Specialization in Hospital Pharmacy, University of Pisa, Pisa, Italy
| | - Andrea Ossato
- Scientific Committee, Italian Society of Clinical Pharmacy and Therapeutics, Turin, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Alessandro Inno
- Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy
| | - Andrea Messori
- Scientific Committee, Italian Society of Clinical Pharmacy and Therapeutics, Turin, Italy
- Health Technology Assessment (HTA) Unit, Regional Health Service, Florence, Italy
| |
Collapse
|
13
|
Goss G, Ciuleanu T, Ramlau R, Renouf DJ, Chu Q, Kalinka E, Sawrycki P, Bramson J, Nelson BH, Crabbé R, LaCasse E, Lo B, Sahlender DA, Crompton P, Brichory F, Piggott L, Schenker M, Juergens R. Xevinapant plus avelumab in advanced solid tumours, with a dose expansion in advanced non-small-cell lung cancer: exploratory biomarker, safety and efficacy analyses from an open-label, nonrandomised phase Ib study. Ther Adv Med Oncol 2025; 17:17588359251332154. [PMID: 40351326 PMCID: PMC12062605 DOI: 10.1177/17588359251332154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/18/2025] [Indexed: 05/14/2025] Open
Abstract
Background Xevinapant, an inhibitor of apoptosis protein (IAP) inhibitor, has shown promising activity in combination with anticancer agents, including radiotherapy, and, in preclinical studies, anti-PD-(L)1 antibodies. This, in part, is due to its ability to restore apoptosis and increase antitumour immunity. Objectives We report efficacy, safety and exploratory biomarker analyses of xevinapant plus avelumab (anti-PD-L1) in a two-part, open-label, nonrandomised, phase Ib study. Design Part A assessed patients with advanced solid tumours who received xevinapant (100, 150, 200 or 250 mg/day, with no random allocation, on Days 1-10 and 15-24) in combination with avelumab (10 mg/kg) on Days 1 and 15 in 28-day cycle. Part B assessed patients with advanced non-small-cell lung cancer (NSCLC) who received xevinapant at the recommended phase II dose (RP2D) plus avelumab (maximum 26 cycles). Methods Part A assessed the safety and tolerability of the combination and established the maximum tolerated dose (MTD) and RP2D of xevinapant. Part B assessed the antitumour activity of xevinapant at the RP2D combined with avelumab compared with a historical control (avelumab alone). Exploratory biomarker analyses were also conducted. Results In part A (n = 16), xevinapant 200 mg/day was established as the RP2D with avelumab and the MTD was not reached. The most common treatment-emergent adverse events (TEAEs) irrespective of xevinapant dose were nausea and fatigue (n = 11 (68.8%) each). In part B (n = 38; four patients received prior anti-PD-(L)1 antibody), the objective response rate (ORR) was 10.5% (95% confidence interval (CI), 2.9-24.8; partial response, n = 4) and the most common TEAE was decreased appetite (n = 13 (34.2%)). Levels of plasma IL-10, IL-1β, IL-13 and CD8+ T cells increased during the study, and circulating levels of CD4+ T cells and Tregs increased during cycle 1. Macrophage-related gene expression signatures increased in patients with a partial response or stable disease. Low baseline Ki-67 expression in tumour samples correlated with a partial response. Conclusion The RP2D of xevinapant with avelumab was established; however, the ORR was not superior to the historical control (avelumab alone). The combination had a manageable safety profile in both study parts. Biomarker analyses provide insights into drivers associated with efficacy in patients with NSCLC receiving xevinapant plus avelumab. Trial registration NCT03270176 (https://clinicaltrials.gov/study/NCT03270176). Registered on ClinicalTrials.gov on 29 August 2017.
Collapse
Affiliation(s)
- Glenwood Goss
- Department of Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H8L6, Canada
| | - Tudor Ciuleanu
- Department of Oncology, ‘Iuliu Haţieganu’ University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Rodryg Ramlau
- Oncology Department, Poznan University of Medical Sciences, Poznan, Poland
| | - Daniel J. Renouf
- Department of Medicine, Faculty of Medicine, University of British Columbia, BC Cancer, Vancouver, BC, Canada
| | - Quincy Chu
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB, Canada
| | - Ewa Kalinka
- Department of Oncology, Polish Mother’s Memorial Hospital, Lodz, Poland
| | - Piotr Sawrycki
- Wojewódzki Szpital Zespolony im. L. Rydygiera w Toruniu, Torun, Poland
| | - Jonathan Bramson
- Centre for Discovery in Cancer Research and Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Brad H. Nelson
- Deeley Research Centre, BC Cancer – Victoria, Victoria, BC, Canada
| | | | | | - Bryan Lo
- Division of Anatomical Pathology, The Ottawa Hospital, Ottawa, ON, Canada
| | | | | | | | - Luke Piggott
- Debiopharm International SA, Lausanne, Switzerland
| | - Michael Schenker
- Oncology Center Sf Nectarie, Craiova, Romania
- Medical Oncology, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Rosalyn Juergens
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
14
|
Gomez-Randulfe I, Monaca F, Planchard D, Bria E, Califano R. Evolving treatment for advanced non-small cell lung cancer harbouring common EGFR activating mutations. Crit Rev Oncol Hematol 2025; 212:104762. [PMID: 40324662 DOI: 10.1016/j.critrevonc.2025.104762] [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: 01/22/2025] [Revised: 04/26/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025] Open
Abstract
A clinically important subgroup of non-small cell lung cancer (NSCLC) is driven by common mutations in the epidermal growth factor receptor (EGFR). Over the past decade, first-, second-, and third-generation EGFR tyrosine kinase inhibitors (TKIs) have substantially improved clinical outcomes, although acquired resistance inevitably emerges. In particular, the third-generation TKI osimertinib has demonstrated superior progression-free survival (PFS) and overall survival (OS) compared to earlier-generation TKIs in the frontline setting, yet median OS remains approximately three years in pivotal trials. Efforts to extend disease control have led to various upfront intensification strategies, including combining EGFR TKIs with antiangiogenics or chemotherapy (e.g., the FLAURA-2 trial), and pairing novel bispecific antibodies such as amivantamab with third-generation TKIs. Upon progression on third-generation EGFR TKIs, platinum-based chemotherapy remains the standard second-line treatment, albeit with modest response rates. Emerging therapies targeting MET amplification (e.g., savolitinib plus osimertinib), leveraging antibody-drug conjugates (e.g., patritumab deruxtecan), or adding immunotherapy and antiangiogenics have shown preliminary promise in overcoming resistance. Ongoing trials are assessing optimal treatment sequencing and the use of circulating tumor DNA (ctDNA) to guide therapy escalation or de-escalation. Ultimately, the evolving landscape of EGFR-mutant NSCLC underscores the need for refined biomarker-driven approaches and personalized regimens to achieve further gains in survival. In this review, we discuss these strategies in detail, highlighting current evidence and future directions for EGFR-mutant NSCLC treatment.
Collapse
Affiliation(s)
- Igor Gomez-Randulfe
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Federico Monaca
- Department of Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - David Planchard
- Department of Medical Oncology, Thoracic Group, Gustave Roussy, Villejuif, France
| | - Emilio Bria
- Università Cattolica del Sacro Cuore, Rome, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Ospedale Isola Tiberina - Gemelli Isola, Rome, Italy
| | - Raffaele Califano
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Division of Cancer Sciences, The University of Manchester, Manchester, UK.
| |
Collapse
|
15
|
Eklund EA, Orgard M, Wallin D, Sayin SI, Fagman H, Isaksson J, Raghavan S, Akyürek LM, Nyman J, Wiel C, Hallqvist A, Sayin VI. Equalizing prognostic disparities in KRAS-mutated stage III NSCLC patients: addition of durvalumab to combined chemoradiotherapy improves survival. Lung Cancer 2025; 204:108573. [PMID: 40349418 DOI: 10.1016/j.lungcan.2025.108573] [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: 09/05/2024] [Revised: 01/28/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
INTRODUCTION Stage III non-small cell lung cancer (NSCLC) is a heterogeneous group and identification of subgroups with differential treatment responses is crucial. Addition of durvalumab to concurrent chemoradiotherapy (cCRT) has previously been shown to improve survival outcomes. Meanwhile, subgroups harboring KRAS mutations have been shown to have worse prognosis. We investigated whether KRAS mutational status may affect survival outcomes after adjuvant durvalumab following cCRT in stage III NSCLC. METHODS In this multi-center retrospective study, we present a real-world dataset of all stage III NSCLC patients treated with curative-intent cCRT with molecular assessment, between 2016 and 2021 in the Västra Götaland Region of western Sweden. The study period includes the standard practice prior to the introduction of durvalumab, enabling evaluation of the potential impact of immune checkpoint blockade (ICB). Primary study outcomes were overall survival (OS) and progression free survival (PFS). RESULTS We identified 145 patients who received cCRT with curative intent, and 32 % harbored an activating mutation in the KRAS gene (KRASMUT; n = 46). Compared to patients with wild-type KRAS (KRASWT; n = 99), KRASMUT had worse OS (p = 0.047) and PFS (p = 0.038). This finding persisted on multivariate analysis with OS (HR 1.703, 95 % CI 1.074-2.702, p = 0.024) and PFS (HR 1.628, 95 % CI 1.081-2.453, p = 0.020). Within the subgroup that received cCRT alone, KRASMUT patients (n = 35) exhibited worse OS (p = 0.036) and PFS (p = 0.037) compared with KRASWT (n = 35). However, among those who received additional durvalumab after cCRT (KRASWT; n = 99. KRASMUT; n = 11) there were no significant differences in OS (0.788) or PFS (0.855) between the groups. CONCLUSIONS KRAS mutations are a negative prognostic factor after cCRT in stage III NSCLC, and the addition of durvalumab ameliorates the negative impact of harboring this mutation.
Collapse
Affiliation(s)
- Ella A Eklund
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden; Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mathilda Orgard
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Delice Wallin
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Sama I Sayin
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Henrik Fagman
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden; Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Johan Isaksson
- Centre of Research and Development Region Gävleborg, Uppsala University, Sweden
| | - Sukanya Raghavan
- Department of Microbiology and Immunology, Institute for Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Levent M Akyürek
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden; Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jan Nyman
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Oncology, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Clotilde Wiel
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
| | - Andreas Hallqvist
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Oncology, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden.
| | - Volkan I Sayin
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
16
|
Tian G, Nie J, Dai L, Hu W, Zhang J, Wu D, Ma X, Chen X, Han S, Han J, Zhang Z, Long J, Zhao X, Fang J. Real-world analysis of the efficacy and safety of lorlatinib in ALK-positive non-small cell lung cancer patients in China. Front Oncol 2025; 15:1577607. [PMID: 40376587 PMCID: PMC12078162 DOI: 10.3389/fonc.2025.1577607] [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: 02/16/2025] [Accepted: 03/31/2025] [Indexed: 05/18/2025] Open
Abstract
Introduction Lorlatinib, a third-generation ALK inhibitor, has demonstrated strong efficacy in treating advanced ALK-positive NSCLC, though real-world data, particularly from China, are limited. This study evaluates the real-world efficacy and safety of lorlatinib in Chinese patients with advanced ALK-positive NSCLC. Materials and methods This retrospective study analyzed 65 patients with advanced ALK-positive NSCLC who received lorlatinib at Peking University Cancer Hospital between September 2017 and August 2024. The study assessed the overall response rate (ORR), progression-free survival (PFS), and safety outcomes, comparing first-line treatment to subsequent treatments after prior ALK inhibitor exposure. Results The real-world ORR (rwORR) for all patients was 49.2%, with a real-world disease control rate (rwDCR) of 92.3%. In the first-line treatment group (n=8), lorlatinib showed an ORR of 100%, and no patients experienced progressive disease (PD) during a median follow-up of 9 months. The mPFS for the entire cohort was 37.83 months, with the median OS (mOS) not reached (NR, 95% CI: NR-NR). Patients who had received one prior ALK inhibitor had a mPFS of 49.73 months, while those who had received two or more prior ALK inhibitors had a mPFS of 12.17 months. A statistically significant difference in mOS was found between patients with one prior ALKi and those with two or more prior ALKis (p = 0.032). Lorlatinib demonstrated strong intracranial efficacy, with a 45.2% intracranial ORR in patients with brain metastases. The safety profile was consistent with previous reports, with the most common AEs being hyperlipidemia. However, the incidence of severe AEs was manageable with dose adjustments and supportive treatments. Conclusions Lorlatinib demonstrates strong efficacy and manageable safety, especially in first-line treatment of advanced ALK-positive NSCLC, supporting its role as an effective treatment option.
Collapse
Affiliation(s)
- Guangming Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Thoracic Oncology Department II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jun Nie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Thoracic Oncology Department II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ling Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Thoracic Oncology Department II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Weiheng Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Thoracic Oncology Department II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jie Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Thoracic Oncology Department II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Di Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Thoracic Oncology Department II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiangjuan Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Thoracic Oncology Department II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaoling Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Thoracic Oncology Department II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Sen Han
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Thoracic Oncology Department II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jindi Han
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Thoracic Oncology Department II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ziran Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Thoracic Oncology Department II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jieran Long
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Thoracic Oncology Department II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xinliang Zhao
- Department of Medical Genetics, School of Basic Sciences, Peking University, Beijing, China
| | - Jian Fang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Thoracic Oncology Department II, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
17
|
Passiglia F, Listì A, Bironzo P, Merlini A, Benso F, Napoli F, Barbu FA, Zambelli V, Tabbò F, Reale ML, Sini C, Roca E, Taveggia PA, Simionato F, Buffoni L, Mazilu L, Barbieri V, Pignataro D, Araujo A, Paz-Ares L, Felip E, Secen N, Comanescu A, Ramizi KM, Bettini AC, Scotti V, Linardou H, Mohorcic K, Meoni G, Giannarelli D, Volante M, Malapelle U, Vallone S, Scagliotti G, Righi L, Novello S. Actionable NSCLC Mutation Identification by Comprehensive Genomic Profiling for Clinical Trial Enrollment: The European Program for the Routine Testing of Patients With Advanced Lung Cancer (EPROPA). J Thorac Oncol 2025; 20:614-624. [PMID: 39694416 DOI: 10.1016/j.jtho.2024.12.010] [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: 06/03/2024] [Revised: 11/15/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024]
Abstract
INTRODUCTION The advocacy Women Against Lung Cancer in Europe (WALCE) promoted the European Program for the Routine Testing of Patients With Advanced Lung Cancer (EPROPA) and provided a free-of-charge molecular profiling platform for NSCLC sample characterization with the aim of increasing the detection of targetable drivers and improving patients' access to clinical trials in Europe. METHODS From January 2021 to December 2023, 20 centers located at five different European countries (Greece, Slovenia, Romania, Albania, and Italy) joined EPROPA, with 555 patients with advanced NSCLC registered to the program. Anonymized patients' clinical-pathological data were shared through the EPROPA web platform and tissue samples were collected at the Molecular Pathology Unit of the Reference Center (University of Turin) for molecular analyses. A comprehensive genomic profiling by a targeted next-generation sequencing approach has been performed and molecular reports have been discussed within the molecular tumor board to assess patients' eligibility for clinical trials. RESULTS The average turnaround time was eight days, with only 30 out of 555 tissue samples (6%) not suitable for molecular analysis. In the 525 analyzed samples, a total of 570 molecular alterations have been identified, including 264 pathogenic targetable oncogenic alterations and 113 cases with co-occurring mutations. A total of 18 molecular alterations with potential germline and hereditary cancer syndrome implications have been reported. The identification of a clinical trial was considered for 205 patients. After molecular tumor board discussion, 30 patients were enrolled and treated in clinical studies available in Europe. Survival outcomes were significantly improved in patients with targetable molecular alterations receiving a matched targeted therapy. CONCLUSION This data confirmed the feasibility and usefulness of the program in the real-world practice scenario, supporting the implementation of next-generation sequencing-based molecular characterization of NSCLC samples, to reduce the unequal access to tests, drugs, and clinical trials in Europe.
Collapse
Affiliation(s)
- Francesco Passiglia
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Angela Listì
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Paolo Bironzo
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Alessandra Merlini
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Federica Benso
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Francesca Napoli
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Francesca Alice Barbu
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Vanessa Zambelli
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Fabrizio Tabbò
- Medical Oncology, ASLCN2 Alba e Bra, Michele e Pietro Ferrero Hospital, Verduno, Cuneo, Italy
| | | | - Claudio Sini
- Medical Oncology and CPDO, Giovanni Paolo II Hospital, Olbia, Italy
| | - Elisa Roca
- Thoracic Oncology - Lung Unit P. Pederzoli Hospital, Peschiera d/G, Verona, Italy
| | | | | | - Lucio Buffoni
- Medical Oncology, Humanitas Gradenigo Hospital, Torino, Italy
| | - Laura Mazilu
- Medical Oncology, Ovidius Clinical Hospital, Ovidius University of Constanta, Romania
| | - Vito Barbieri
- Medical Oncology, Renato Dulbecco University Hospital, Catanzaro, Italy
| | | | - Antonio Araujo
- Department of Medical Oncology, Centro Hospitalar Universitário de Santo António, Portugal
| | - Luis Paz-Ares
- Medical Oncology Department, University Hospital 12 De Octubre, Madrid, Spain
| | - Enriqueta Felip
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Nevena Secen
- Pulmonology-Oncology Department, AcibademBelmedic, Belgrade, Serbia
| | | | | | | | - Vieri Scotti
- Department of Radiotherapy, AOU Careggi, Firenze, Italy
| | - Helena Linardou
- Fourth Oncology Department and Comprehensive Clinical Trials Center, Metropolitan Hospital, Athens, Greece
| | - Katja Mohorcic
- Medical Oncology Department, University Clinic of Respiratory and Allergic Diseases, Golnik, Slovenia
| | - Giulia Meoni
- SOS Medical Oncology, San Giovanni di Dio Hospital, Firenze, Italy
| | - Diana Giannarelli
- Fondazione Policlinico Universitario A. Gemelli, IRCCS - Facility of Epidemiology and Biostatistics, Rome, Italy
| | - Marco Volante
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | | | - Giorgio Scagliotti
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Luisella Righi
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy
| | - Silvia Novello
- Department of Oncology, University of Turin, S. Luigi Gonzaga Hospital, Orbassano, Turin, Italy.
| |
Collapse
|
18
|
Wu Q, Li Q, Qin Y. A cost-effectiveness analysis of amivantamab plus lazertinib versus osimertinib in the treatment of US and Chinese patients with EGFR-mutated advanced non-small cell lung cancer. Lung Cancer 2025; 203:108533. [PMID: 40220717 DOI: 10.1016/j.lungcan.2025.108533] [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: 02/07/2025] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND The combination of amivantamab and lazertinib (AL) has demonstrated clinically significant efficacy in patients with previously untreated EGFR-mutated advanced non-small cell lung cancer (NSCLC). However, its economic value relative to the standard therapy, osimertinib, remains unclear. This study evaluates the cost-effectiveness of AL regimen compared with osimertinib in US and Chinese healthcare settings. METHODS A partitioned survival model, comprising progression-free survival (PFS), post-progression, and death states, was developed using a Markov model. Clinical data were obtained from the recent Phase III MARIPOSA trial. Direct medical costs (including drug acquisition, administration, and adverse event management) were obtained from US and Chinese healthcare system data, public databases, and the literature. Health-state utilities were sourced from the literature. Incremental cost-effectiveness ratios (ICERs) were calculated based on quality-adjusted life years (QALYs). Threshold analysis was performed to identify pricing strategies at specified willingness-to-pay (WTP) thresholds. Model robustness was assessed through sensitivity and scenario analyses, and additional subgroup analyses performed. RESULTS In the base case analysis, the average costs of AL and osimertinib regimen were $1,030,524.3 (China: $234,270.87) and $466,922.0 (China: $20,075.35), respectively, and the QALYs achieved were 4.08 (China: 3.66) and 2.60 (China: 2.66), respectively. The ICERs for AL compared with osimertinib in the US and China were $563,602.3 and $214,195.51, respectively. Based on the respective WTP thresholds in the US and China, the AL regimen did not represent a cost-effective option. Sensitivity, scenario, and subgroup analyses confirmed the robustness of these findings. CONCLUSIONS Although AL regimen prolongs QALYs compared with osimertinib, it may not meet cost-effectiveness thresholds given current US pricing and simulated Chinese prices. These findings emphasize the need to consider policy implications and future pricing strategies.
Collapse
Affiliation(s)
- Qiuji Wu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, China
| | - Qiu Li
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, China
| | - Yi Qin
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
19
|
Zhou X, Zeng L, Huang Z, Ruan Z, Yan H, Zou C, Xu S, Zhang Y. Strategies Beyond 3rd EGFR-TKI Acquired Resistance: Opportunities and Challenges. Cancer Med 2025; 14:e70921. [PMID: 40322930 PMCID: PMC12051098 DOI: 10.1002/cam4.70921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
The seminal identification of epidermal growth factor receptor (EGFR) mutations as pivotal oncogenic drivers in non-small cell lung cancer (NSCLC) has catalyzed the evolution of biomarker-guided therapeutic paradigms for advanced disease. Currently, third-generation EGFR tyrosine kinase inhibitors (EGFR-TKI) have revolutionized first-line treatment for advanced EGFR-mutated NSCLC, yet acquired resistance remains an inevitable and formidable clinical challenge. This review systematically summarizes molecular mechanisms underlying treatment resistance, with a focus on clinical challenges associated with central nervous system (CNS) metastases. Therapeutic resistance mechanisms are categorized into EGFR-dependent (on-target) pathways, typified by acquired kinase domain mutations (e.g., C797S), and EGFR-independent (off-target) pathways, involving compensatory activation of parallel signaling effectors (e.g., MET amplification, HER2 activation) or phenotypic transformation. We further evaluated contemporary diagnostic modalities for identifying resistance drivers and appraised emerging therapeutic strategies, including fourth-generation EGFR-TKI, various combination therapies, and antibody-drug conjugates (ADCs), and so forth, with emphasis on ongoing clinical trials that may transform the existing treatment paradigm. By synthesizing preclinical and clinical insights, this review aims to advance mechanistic understanding and propose therapeutic strategies to overcome acquired resistance to third-generation EGFR-TKI in first-line treatment.
Collapse
Affiliation(s)
- Xuexue Zhou
- Medical CollegeJishou UniversityJishouChina
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Liang Zeng
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Zhe Huang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
- Department of Pathology and Pathophysiology, School of Basic Medical SciencesCentral South UniversityChangshaChina
| | - Zhaohui Ruan
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
- Department of Pathology and Pathophysiology, School of Basic Medical SciencesCentral South UniversityChangshaChina
| | - Huan Yan
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Chun Zou
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Shidong Xu
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
- Department of Pathology and Pathophysiology, School of Basic Medical SciencesCentral South UniversityChangshaChina
| | - Yongchang Zhang
- Medical CollegeJishou UniversityJishouChina
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
- Department of Pathology and Pathophysiology, School of Basic Medical SciencesCentral South UniversityChangshaChina
| |
Collapse
|
20
|
Wong WKY, Mok KKS, Tsui GPC, Chen OH, Loong HHF, Chan LL, Yan K, Mo F, Lee KWC, Lam KC, Mok FST, Johnson D, Chen ACC, Lam B, Lee M, Mok TSK, Li MSC. CNS Outcomes of Osimertinib Plus Chemotherapy in Patients With EGFR Mutation Positive Lung Cancer Beyond Osimertinib Progression. Clin Lung Cancer 2025; 26:e214-e222.e5. [PMID: 39933981 DOI: 10.1016/j.cllc.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Central nervous system (CNS) metastases are common among patients with epidermal growth factor receptor (EGFR) mutation positive non-small cell lung cancer (NSCLC). Osimertinib in combination with chemotherapy beyond osimertinib progression may minimize CNS progression. METHOD In this retrospective analysis, patients with advanced EGFR mutation positive NSCLC and brain metastases who received platinum-based chemotherapy (PbChT) after disease progression on osimertinib were enrolled. The primary endpoint was real-world CNS progression-free survival (rwCNS-PFS) between patients who received PbChT with and without osimertinib continuation. Secondary endpoints included competing risk analysis of CNS progression and incidence of salvage radiotherapy to brain. RESULTS A total of 101 patients were analyzed, out of which, 39 (39%) continued osimertinib with chemotherapy (OSI+ cohort) and 62 (61%) received chemotherapy alone (OSI- cohort). Median rwCNS-PFS was significantly longer in the OSI+ cohort (9.0 months, 95% CI 6.6-11.4) than the OSI- cohort (5.7 months, 95% CI 4.6-6.9) (HR 0.37, 95% CI 0.18-0.76, P = .007). This remained significant after adjustment for EGFR mutation, line of osimertinib treatment, prior radiotherapy to brain, and CNS progression on osimertinib monotherapy. Estimated probability of CNS progression at 6 months was 5.6% in OSI+ cohort versus 20.9% in OSI- cohort. Incidence of salvage radiotherapy to brain was lower in the OSI+ cohort (15%) compared to OSI- cohort (24%). CONCLUSION In patients with EGFR mutation positive NSCLC and brain metastases, continuing osimertinib with chemotherapy after progression on osimertinib significantly reduced risk of CNS progression. Prospective studies are warranted to define the optimal treatment strategy for this patient population.
Collapse
Affiliation(s)
- Wesley K Y Wong
- Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, China
| | - Kevin K S Mok
- Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, China
| | - Giselle P C Tsui
- Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, China
| | - Olivia H Chen
- Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, China
| | - Herbert H F Loong
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Landon L Chan
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Kelvin Yan
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Frankie Mo
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Kirsty W C Lee
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - K C Lam
- Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, China
| | - Florence S T Mok
- Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, China
| | - David Johnson
- Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, China
| | - Allen C C Chen
- Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, China
| | - Benjamin Lam
- Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, China
| | - Matthew Lee
- Department of Clinical Oncology, Prince of Wales Hospital, Hong Kong, China
| | - Tony S K Mok
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China
| | - Molly S C Li
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
21
|
Heersche N, Lanser DAC, Oomen-de Hoop E, Içli A, de Bruijn P, Paats MS, van Rossum EFC, Koolen SLW, van Schaik RHN, Dingemans AMC, Veerman GDM, Mathijssen RHJ. Semaglutide impairs bioavailability of alectinib: a note of warning based on a cross-over pharmacokinetic drug-drug interaction study. Cancer Commun (Lond) 2025. [PMID: 40308149 DOI: 10.1002/cac2.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 04/14/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025] Open
Affiliation(s)
- Niels Heersche
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Daan A C Lanser
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Pulmonology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Esther Oomen-de Hoop
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Attila Içli
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Peter de Bruijn
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marthe S Paats
- Department of Pulmonology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Stijn L W Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Pharmacy, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Anne-Marie C Dingemans
- Department of Pulmonology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - G D Marijn Veerman
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Pulmonology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
22
|
Artal-Cortes Á, Gimeno-Pelegrín J. Osimertinib and Performance Status 2: Not Every Patient Fits Into a Pivotal Clinical Trial. J Thorac Oncol 2025; 20:557-559. [PMID: 40348481 DOI: 10.1016/j.jtho.2025.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 05/14/2025]
Affiliation(s)
- Ángel Artal-Cortes
- Servicio de Oncología Médica, Hospital Universitario Miguel Servet, Zaragoza, Spain.
| | | |
Collapse
|
23
|
Rapa I, Bertola F, Roversi G, Seminati D, Panebianco F, Durães C, Gallo E, Leone BE, Palange A, Righi L, Visca P, Volante M, Buglioni S. Impact and Reproducibility of In-House Targeted Next-Generation Sequencing Biomarker Testing in Non-Small-Cell Lung Cancer: An Italian Multi-Institutional Experience. J Mol Diagn 2025; 27:371-382. [PMID: 40023494 DOI: 10.1016/j.jmoldx.2025.02.001] [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: 01/15/2024] [Revised: 11/14/2024] [Accepted: 02/05/2025] [Indexed: 03/04/2025] Open
Abstract
Next-generation sequencing (NGS) allows the detection of multiple genetic targets in different tumor types. This study aimed to confirm the benefits of implementing in-house NGS testing for non-small-cell lung cancer (NSCLC) samples in molecular pathology laboratories. A multi-institutional study was conducted to evaluate the analytical performance, turnaround time, and feasibility of in-house NGS testing of 50 genes from 283 NSCLC samples. The first phase was a retrospective study with interlaboratory testing (21 samples), and the second phase was a prospective study with intralaboratory testing (262 samples). The retrospective study showed a 100% sequencing success rate for DNA and RNA, high interlaboratory concordance (95.2%), and a strong correlation (R2 = 0.94) between observed and expected single-nucleotide variant/insertion and/or deletion variant allele fraction. The prospective study showed a sequencing success rate of 99.2% for DNA and 98% for RNA. NGS identified 285 relevant variants (81.1% single-nucleotide variants/insertion and/or deletion variants, 9.8% copy number variants, and 9.1% gene fusions). Co-mutations with potential clinical relevance were detected in 20.5% of samples positive for the main oncogenic drivers in NSCLC. Additionally, 11% of samples wild type for the main oncogenic drivers carried alterations in other relevant genes. The in-house NGS testing had a median turnaround time from sample processing to molecular report of 4 days. This study demonstrates the advantages of implementing in-house NGS testing in molecular pathology laboratories.
Collapse
Affiliation(s)
- Ida Rapa
- Pathology Unit, San Luigi Gonzaga Hospital, Turin, Italy
| | - Francesca Bertola
- Medical Genetics Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Gerardo dei Tintori, Monza, Italy
| | - Gaia Roversi
- Medical Genetics Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Gerardo dei Tintori, Monza, Italy; Department of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Davide Seminati
- Oncological Molecular Pathology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Federica Panebianco
- Medical and Scientific Affairs, Thermo Fisher Scientific, Waltham, Massachusetts
| | - Cecília Durães
- Medical and Scientific Affairs, Thermo Fisher Scientific, Waltham, Massachusetts
| | - Enzo Gallo
- Department of Pathology, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Biagio E Leone
- Department of Medicine and Surgery, University of Milano Bicocca, Milan, Italy; Oncological Molecular Pathology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Aldo Palange
- Department of Pathology, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Luisella Righi
- Department of Oncology, University of Turin, Turin, Italy
| | - Paolo Visca
- Department of Pathology, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Marco Volante
- Department of Oncology, University of Turin, Turin, Italy
| | - Simonetta Buglioni
- Department of Pathology, IRCCS, Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
24
|
Chouaid C, Bosquet L, Knott C, Li Z, Schaeffer M, Lin X, Schioppa CA, Perualila NJ, Diels J, Caparros EQ, Galea F, Hultén A, Greystoke A. Real-world frontline treatments in patients with advanced non-small-cell lung cancer harboring epidermal growth factor receptor exon 20 insertions and adjusted comparisons versus amivantamab plus chemotherapy from the PAPILLON study. Lung Cancer 2025; 203:108548. [PMID: 40262227 DOI: 10.1016/j.lungcan.2025.108548] [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: 12/06/2024] [Revised: 04/13/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025]
Abstract
INTRODUCTION In PAPILLON, frontline amivantamab + carboplatin + pemetrexed (ACP) demonstrated superior efficacy over carboplatin + pemetrexed in patients with advanced or metastatic non-small-cell lung cancer (aNSCLC) harboring mutations in epidermal growth factor receptor (EGFR) exon 20 insertions (exon20ins). Real-world (RW) treatment patterns and comparative effectiveness of ACP versus RW treatments are unknown. MATERIALS AND METHODS The present study (NECTAR) retrospectively analyzed frontline treatments prescribed 2012-2023 for patients with aNSCLC and confirmed EGFR exon20ins from English (ENG-NCRD), French (FR-ESME), and US (US-COTA and US-ConcertAI) datasets. Overall survival (OS), time to next treatment (TTNT), and progression-free survival (PFS) were assessed in RW pooled and individual treatment classes and in indirect treatment comparisons (ITC) between ACP from PAPILLON and RW treatments using Cox proportional hazards model adjusted for prognostic factors. RESULTS NECTAR assessed 208 RW patients: ENG-NCRD, n = 23; FR-ESME, n = 91; US-COTA, n = 39, and US-ConcertAI, n = 55. Common frontline treatment classes were platinum-based chemotherapy (33.7 %), platinum + immunotherapy (23.1 %), EGFR tyrosine kinase inhibitors (TKIs) alone (15.4 %), platinum + VEGF inhibitors (VEGFi) (11.1 %), and immunotherapy alone (7.7 %). Compared with platinum-based chemotherapy, none of the evaluated treatment classes demonstrated improved OS, TTNT, and PFS. Exceptions were platinum + VEGFi in TTNT and PFS and platinum + immunotherapy in TTNT. In ITCs, ACP significantly improved OS over pooled RW treatments (HR = 0.48 [95 % CI, 0.32-0.71]; P < 0.001), platinum-based chemotherapy (HR = 0.48 [0.30-0.77]; P = 0.003), platinum + immunotherapy (HR = 0.41 [0.23-0.73]; P = 0.003), and EGFR TKI alone (HR = 0.48 [0.23-1.02]; P = 0.055). TTNT and PFS results were similar to OS. CONCLUSIONS In patients with EGFR exon20ins aNSCLC, frontline ACP was superior to common RW treatments, highlighting the need for practice change.
Collapse
Affiliation(s)
| | - Lise Bosquet
- Unicancer, 101 rue de Tolbiac, 75013 Paris, France
| | - Craig Knott
- Health Data Insight CIC, CPC4,Capital Park, Cambridge CB2 15XE, UK; National Disease Registration Service, NHS England, 10 South Colonnade, Canary Wharf, London E14 4PU, UK
| | - Ziming Li
- Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai, China
| | | | - Xiwu Lin
- Johnson & Johnson, 850 Ridgeview Dr, Horsham, PA, USA
| | | | | | - Joris Diels
- Johnson & Johnson, Turnhoutseweg 30, 2340 Beerse, Belgium
| | | | - Francesca Galea
- Johnson & Johnson, 50-100 Holmers Farm Way, High Wycombe, UK
| | - Annika Hultén
- Johnson & Johnson, Hatsinanpuisto 8B, PL 15, 02621 Espoo, Finland
| | | |
Collapse
|
25
|
Stanzione B, Del Conte A, Bertoli E, De Carlo E, Bortolot M, Torresan S, Spina M, Bearz A. Non-Small Cell Lung Cancer with Epidermal Growth Factor Receptor (EGFR) Common Mutations: New Strategies. Cancers (Basel) 2025; 17:1515. [PMID: 40361442 PMCID: PMC12071048 DOI: 10.3390/cancers17091515] [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: 02/11/2025] [Revised: 04/23/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
Epidermal growth factor receptor (EGFR) mutations are described in 10-15% of Caucasian patients and in 50% of Asian patients with non-squamous non-small cell lung cancer (NSCLC). The introduction of tyrosine kinase inhibitors (TKIs) has revolutionized the therapeutic scenario and has changed the natural history of the disease. Despite the results obtained with osimertinib, a third-generation TKI, most patients experience disease progression. The search for new therapeutic strategies both to enhance first-line treatment and to ensure adequate second-line therapies represents an unmet medical need, towards which all efforts are being concentrated. In this review, we describe the main strategies identified to improve the prognosis of patients with EGFR-mutated NSCLC.
Collapse
Affiliation(s)
- Brigida Stanzione
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (A.D.C.); (E.B.); (E.D.C.); (M.B.); (S.T.); (M.S.); (A.B.)
| | - Alessandro Del Conte
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (A.D.C.); (E.B.); (E.D.C.); (M.B.); (S.T.); (M.S.); (A.B.)
| | - Elisa Bertoli
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (A.D.C.); (E.B.); (E.D.C.); (M.B.); (S.T.); (M.S.); (A.B.)
| | - Elisa De Carlo
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (A.D.C.); (E.B.); (E.D.C.); (M.B.); (S.T.); (M.S.); (A.B.)
| | - Martina Bortolot
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (A.D.C.); (E.B.); (E.D.C.); (M.B.); (S.T.); (M.S.); (A.B.)
- Department of Medicine (DME), University of Udine, 33100 Udine, Italy
| | - Sara Torresan
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (A.D.C.); (E.B.); (E.D.C.); (M.B.); (S.T.); (M.S.); (A.B.)
- Department of Medicine (DME), University of Udine, 33100 Udine, Italy
| | - Michele Spina
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (A.D.C.); (E.B.); (E.D.C.); (M.B.); (S.T.); (M.S.); (A.B.)
| | - Alessandra Bearz
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (A.D.C.); (E.B.); (E.D.C.); (M.B.); (S.T.); (M.S.); (A.B.)
| |
Collapse
|
26
|
Serna-Blasco R, Mediavilla-Medel P, Medina K, Sala MÁ, Aguiar D, Díaz-Serrano A, Antoñanzas M, Ocaña J, Mielgo X, Fernández I, López-Castro R, Cobo M, Martínez M, Villa JC, Rosado P, López A, Guirado M, Viteri S, Rodríguez D, García F, Simón S, Moreno MÁ, Catot S, González Larriba JL, Salas C, Calvo V, Romero A, Provencio M. Comprehensive molecular profiling of advanced NSCLC using NGS: Prevalence of druggable mutations and clinical trial opportunities in the ATLAS study. Lung Cancer 2025; 204:108550. [PMID: 40300279 DOI: 10.1016/j.lungcan.2025.108550] [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: 02/19/2025] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 05/01/2025]
Abstract
BACKGROUND In Spain, next-generation sequencing (NGS) is currently available in a limited number of specialized centers and remains inaccessible to a significant proportion of patients. The ATLAS study aims to explore the tumor molecular profile beyond known EGFR mutations and ALK translocations using NGS on tumor biopsy samples. METHODS Patients with EGFR-sensitizing mutations or ALK translocations were excluded. A total of 455 patients with advanced non-small cell lung cancer (NSCLC) were enrolled from 22 Spanish hospitals. DNA and RNA were extracted from formalin-fixed paraffin-embedded samples, and libraries were prepared using the Oncomine Focus Assay. The Trialing app was used to identify active clinical trials in Spain. RESULTS Mutations were detected in 65.7 % of the cases. Local pathology assessments detected druggable mutations in only 7.9 % of cases, while centralized NGS testing increased this detection rate to 25.9 %. The most prevalent druggable alteration was KRAS G12C (53.6 %), followed by MET amplification (8.1 %) and MET exon 14 skipping (7.3 %). Additionally, 34.5 % of patients had molecular alterations matching clinical trials, offering potential treatment opportunities. Women had a significantly higher probability of harbouring druggable mutations (36 % vs. 20.3 %, p < 0.001), including the KRAS G12C which was significantly more frequent in females (22.6 % vs. 10 %). KRAS mutations were more common in adenocarcinomas and increased with tumor differentiation grade (p < 0.001 and p = 0.049, respectively). Likewise, ALK translocations, EGFR mutations, BRAF V600E, MET exon 14 skipping, and RET/ROS1 fusions were mainly found in adenocarcinomas whereas copy number variations were more frequent in squamous carcinomas (28.6 % vs. 15.1 %; p = 0.003) and in men (22 % vs. 11.6 %; p = 0.008). CONCLUSION The ATLAS study demonstrates the utility of comprehensive NGS testing, which detects clinically relevant mutations in more than one-third of patients and may extend therapy options.
Collapse
Affiliation(s)
- Roberto Serna-Blasco
- Liquid Biopsy Laboratory, Medical Oncology Department. Instituto de Investigación Biomédica Puerta de Hierro - Segovia de Arana, Majadahonda, Comunidad de Madrid, Spain
| | - Pilar Mediavilla-Medel
- Liquid Biopsy Laboratory, Medical Oncology Department. Instituto de Investigación Biomédica Puerta de Hierro - Segovia de Arana, Majadahonda, Comunidad de Madrid, Spain
| | - Karla Medina
- Department of Oncology, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Islas Canarias, Spain
| | - María Ángeles Sala
- Department of Oncology, Hospital Universitario Basurto, Bilbao, País Vasco, Spain
| | - David Aguiar
- Department of Oncology, Hospital Universitario de Gran Canaria Doctor Negrín, Las Palmas de Gran Canaria, Islas Canarias, Spain
| | - Asunción Díaz-Serrano
- Complejo Público Asistencial de Zamora, Hospital Virgen de la Concha, Zamora, Castilla y León, Spain
| | - Mónica Antoñanzas
- Department of Oncology, Hospital Universitario Clínico San Carlos, Madrid, Comunidad de Madrid, Spain
| | - Julio Ocaña
- Department of Oncology, Hospital Sanitas CIMA, Barcelona, Cataluña, Spain
| | - Xabier Mielgo
- Department of Oncology, Hospital Universitario Fundación Alcorcón, Alcorcón, Comunidad de Madrid, Spain
| | - Inmaculada Fernández
- Department of Oncology, Hospital Universitario Virgen Macarena, Sevilla, Andalucía, Spain
| | - Rafael López-Castro
- Department of Oncology, Hospital Clínico Universitario de Valladolid, Valladolid, Castilla y León, Spain
| | - Manuel Cobo
- Department of Oncology, Hospital Regional Universitario de Málaga, Málaga, Andalucía, Spain
| | - Mireia Martínez
- Department of Oncology, Hospital Universitario de Álava - Txagorritxu, Vitoria, País Vasco, Spain
| | - José Carlos Villa
- Department of Oncology, Hospital General Universitario de Ciudad Real, Ciudad Real, Castilla-La Mancha, Spain
| | - Petra Rosado
- Department of Oncology, Hospital Universitario Puerto Real, Puerto Real, Andalucía, Spain
| | - Ana López
- Department of Oncology, Hospital Universitario Severo Ochoa, Leganés, Comunidad de Madrid, Spain
| | - María Guirado
- Department of Oncology, Hospital General Universitario de Elche, Elche, Conunidad Valenciana, Spain
| | - Santiago Viteri
- UOMI Cancer Center, Clínica Mis Tres Torres Barcelona, Cataluña, Spain
| | - Delvys Rodríguez
- Department of Oncology, Hospital Universitario Insular de Gran Canaria, Islas Canarias, Spain
| | - Florencia García
- Department of Oncology, Hospital Quirónsalud Barcelona, Barcelona, Cataluña, Spain
| | - Soraya Simón
- Department of Oncology, Hospital Virgen de la Luz, Cuenca, Castilla-La Mancha, Spain
| | - María Ángeles Moreno
- Department of Oncology, Hospital Universitario De Jerez, Jerez de la Frontera, Andalucía, Spain
| | | | | | - Clara Salas
- Pathology Department, Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | - Virginia Calvo
- Department of Oncology Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | - Atocha Romero
- Liquid Biopsy Laboratory, Medical Oncology Department. Instituto de Investigación Biomédica Puerta de Hierro - Segovia de Arana, Majadahonda, Comunidad de Madrid, Spain; Department of Oncology Hospital Universitario Puerta de Hierro, Majadahonda, Spain.
| | - Mariano Provencio
- Department of Oncology Hospital Universitario Puerta de Hierro, Majadahonda, Spain.
| |
Collapse
|
27
|
Akkus E, Öksüz NE, Erul E. KRAS G12C inhibitors as monotherapy or in combination for metastatic colorectal cancer: A proportion and comparative meta-analysis of efficacy and toxicity from phase I-II-III trials. Crit Rev Oncol Hematol 2025; 211:104741. [PMID: 40274247 DOI: 10.1016/j.critrevonc.2025.104741] [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: 02/19/2025] [Revised: 03/26/2025] [Accepted: 04/19/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND 1-2 % of metastatic colorectal cancers (mCRC) harbor an activating KRAS-G12C mutation. This study aims to pool the results of available clinical trials of KRAS-G12C inhibitors, comparing monotherapy and combinations. METHODS A systematic literature search was conducted in the MEDLINE database and ESMO/ASCO meeting abstracts. Phase I-II-III trials that investigated a KRAS-G12C inhibitor in patients with mCRC were included. The primary endpoints were objective response rate (ORR) and progression-free survival (PFS). Pooled proportions and comparative subgroup analyses for monotherapy and combinations were presented with the random effects model. RESULTS 596 patients with previously treated mCRC in 14 study cohorts treated with one of sotorasib, adagrasib, divarasib, or olomorasib as monotherapy or in combination with cetuximab/panitumumab were included. Combination treatment revealed an ORR of 33.9 % (95 %CI: 20.7-48.4) (I2: 87.1), which is significantly higher than monotherapy [16.7 %, (95 %CI: 8.3-27.3) (I2: 73.2)] (p = 0.045). Median PFS was significantly longer with the combination [5.7 months (95 %CI: 4.4-7.1) (I2: 80.8) vs. 4.2 months (95 %CI: 3.6-4.7) (I2:0.0), p = 0.027]. Grade 3-4 treatment-related adverse events (TRAEs) were significantly more frequent with the combination [32.8 % (95 %CI: 26.4-39.6) (I2:42.5) vs.16.5 % (95 %CI: 4.9-33.1) (I2: 84.2), p = 0.047]. Common adverse events specific to the combinations were skin toxicities, paronychia, and hypomagnesemia. CONCLUSION This analysis suggests that KRAS-G12C inhibitors in combination with anti-EGFR agents may provide a doubled ORR and 1.5-month PFS benefit compared to monotherapy in previously treated mCRC patients, but with a doubled grade 3-4 TRAEs, including skin toxicities, paronychia, and hypomagnesemia. Treatment preferences should be individualized in these highly pretreated patients.
Collapse
Affiliation(s)
- Erman Akkus
- Ankara University Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye; Ankara University Cancer Research Institute, Ankara, Türkiye.
| | - Nejat Emre Öksüz
- Ankara University Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye; Ankara University Cancer Research Institute, Ankara, Türkiye
| | - Enes Erul
- Ankara University Faculty of Medicine, Department of Medical Oncology, Ankara, Türkiye; Ankara University Cancer Research Institute, Ankara, Türkiye
| |
Collapse
|
28
|
de Castro J, Alonso-Fernández P, Castrodeza JJ, Gayete Á, Hernando F, Martínez-Olmos J, Massuti B, Paz-Ares L, Sisó-Almirall A, Vicente D, Molins L. Monitoring of the oncological process for lung cancer in Spain: an expert consensus report. Clin Transl Oncol 2025:10.1007/s12094-025-03883-4. [PMID: 40261489 DOI: 10.1007/s12094-025-03883-4] [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: 12/04/2024] [Accepted: 02/19/2025] [Indexed: 04/24/2025]
Abstract
INTRODUCTION Continuous monitoring of the oncological process is essential for identifying inefficiencies and areas of improvement, enabling better resource allocation in the care of lung cancer patients. OBJECTIVE The objective is to define key indicators and identify critical variables for monitoring lung cancer care, aiming to improve early detection, reduce delays in diagnosis and treatment, and enhance biomarker research, ensuring timely and effective treatments for all patients. METHODS A multidisciplinary expert group conducted a consensus process based on a review of national guidelines and initiatives related to lung cancer care. The experts defined relevant indicators and identified variables for monitoring overall care, addressing delays, and improving biomarker research. The feasibility of incorporating these indicators into existing information systems was also assessed. RESULTS The proposed indicators provide a structured approach for assessing lung cancer care and outcomes. Their inclusion in healthcare information systems would improve the monitoring and evaluation of care quality and patient outcomes. Additionally, these indicators would also promote interoperability and continuous patient care across different centers and regions, allowing informed decision-making in the improvement of healthcare processes by those responsible for healthcare management. CONCLUSIONS The adoption of standardized indicators for lung cancer care monitoring can drive continuous improvement in healthcare processes. Implementing these indicators in information systems will enable better resource allocation, timely and effective treatment, and enhanced coordination among healthcare providers, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Javier de Castro
- Department of Medical Oncology, Hospital Universitario La Paz, IdiPaz, Madrid, Spain.
| | | | - José Javier Castrodeza
- Department of Preventive Medicine, Hospital Clínico Universitario de Valladolid, Castilla y León, Valladolid, Spain
| | - Ángel Gayete
- Departament of Diagnostic Radiology, Hospital del Mar de Barcelona, Cataluña, Barcelona, Spain
| | | | | | - Bartomeu Massuti
- Department of Medical Oncology, Hospital General Universitario de Alicante, Valencia, Spain
| | - Luis Paz-Ares
- Department of Medical Oncology, Hospital Universitario 12 Octubre, Madrid, Spain
| | - Antoni Sisó-Almirall
- Consorci d'Atenció Primària de Salut Barcelona Esquerra (CAPSBE), Cataluña, Barcelona, Spain
| | - David Vicente
- Department of Medical Oncology, Hospital Universitario Virgen Macarena, Sevilla, Andalucía, Spain
| | - Laureano Molins
- Department of Thoracic Surgery, Hospital Clínic, Barcelona University, Cataluña, Barcelona, Spain
| |
Collapse
|
29
|
Rodríguez González M, Montero González JC, Sayagués Manzano JM, Sánchez TC, Ruiz JR, Iglesias Heras M, Rivas Marcos MB, Abad Hernández M, Cordovilla Pérez R. High-Quality Samples for Next-Generation Sequencing and PD-L1 Assessment in Non-Small Cell Lung Cancer: The Role of Endobronchial Ultrasound-Guided Transbronchial Needle Aspiration. Diagnostics (Basel) 2025; 15:1064. [PMID: 40361881 PMCID: PMC12071477 DOI: 10.3390/diagnostics15091064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/04/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: Recent advances in the treatment of non-small cell lung cancer (NSCLC) have shifted from conventional chemotherapy to targeted therapies aimed at specific genetic mutations, particularly in the adenocarcinoma subtype. These therapies have improved overall survival and quality of life. However, some patients still face barriers to accessing these treatments due to challenges in diagnosing advanced-stage NSCLC. Limited tumor cellularity in small biopsies and cytological samples hinders the ability to perform further molecular analyses. Additionally, the increasing number of genetic alterations requiring testing complicates the diagnostic process. To overcome this challenge, we propose combining endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) with next-generation sequencing (NGS) and immunohistochemistry for PD-L1. Methods: A total of 120 EBUS-TBNA samples were consecutively collected during the first year of integrating NGS at a reference hospital in Castilla y León, Spain. Depending on the histology and patient characteristics, a total of 67 NGS analyses and 116 PD-L1 determinations were performed. Results: The cytological sample obtained in these cases successfully achieved the triple objective proposed by the NCCN for lung cancer (diagnosis, staging, and molecular analysis in a single procedure) in 97% of instances. Conclusions: Our study highlights the effectiveness of EBUS-TBNA as a comprehensive, cost-effective, and safe diagnostic tool for NSCLC, successfully achieving the triple objective of diagnosis, staging, and molecular analysis in 97% of cases. The procedure consistently provided high-quality samples for NGS and PD-L1 testing, with minimal complications, reinforcing its value as a reliable approach for optimizing personalized treatment strategies.
Collapse
Affiliation(s)
- Marta Rodríguez González
- Department of Pathology, Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca, 37007 Salamanca, Spain; (J.C.M.G.); (J.M.S.M.)
| | - Juan Carlos Montero González
- Department of Pathology, Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca, 37007 Salamanca, Spain; (J.C.M.G.); (J.M.S.M.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | - José María Sayagués Manzano
- Department of Pathology, Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca, 37007 Salamanca, Spain; (J.C.M.G.); (J.M.S.M.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | | | - Jonnathan Roldán Ruiz
- Department of Clinical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain
| | | | - María Belén Rivas Marcos
- Department of Pathology, Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca, 37007 Salamanca, Spain; (J.C.M.G.); (J.M.S.M.)
| | - Mar Abad Hernández
- Department of Pathology, Biomedical Research Institute of Salamanca (IBSAL), University Hospital of Salamanca, 37007 Salamanca, Spain; (J.C.M.G.); (J.M.S.M.)
| | | |
Collapse
|
30
|
Murat-Onana ML, Ramalingam SS, Jänne PA, Gray JE, Ahn MJ, John T, Yatabe Y, Huang X, Rukazenkov Y, Javey M, Brown H, Li-Sucholeiki X. EGFR mutation testing across the osimertinib clinical program. Lung Cancer 2025; 204:108549. [PMID: 40311309 DOI: 10.1016/j.lungcan.2025.108549] [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: 10/07/2024] [Revised: 03/06/2025] [Accepted: 04/18/2025] [Indexed: 05/03/2025]
Abstract
OBJECTIVES EGFR-tyrosine kinase inhibitors, including osimertinib, have revolutionized EGFR-mutated non-small cell lung cancer (NSCLC) treatment; therefore, early identification of EGFR mutations is essential. We report post-hoc analyses of pooled EGFR mutation tissue testing across osimertinib clinical trials, highlighting testing challenges and supporting best practice. MATERIALS AND METHODS Pooled central Cobas® EGFR Mutation Test data from nine global osimertinib NSCLC clinical trials were analyzed by specimen type, disease stage, and geographical region for specimen adequacy for testing and valid test results. RESULTS Across 4,864 biopsies and 2,402 resections, 91% were adequate for testing, of which 95% of biopsies and 99% of resections had valid test results. Of biopsies, 12% were inadequate for testing (mainly due to insufficient tumor content [42%] and insufficient tissue volume [35%]) and 3% of resections were inadequate (insufficient tumor content [55%] and incorrect specimen preparation [12%]). Inadequacy varied by disease stage, from 3% in resectable stage IA2-IIIA to 10%-15% in first and second/later-line advanced/metastatic settings, and 16% in unresectable stage III. Test success rates among adequate specimens ranged from 93% (unresectable stage III) to 99% (resectable stage IA2-IIIA). Data were similar by geography. DISCUSSION Most tissue specimens were adequate for EGFR testing. Inadequacy was commonly due to insufficient tissue volume or tumor content and higher in biopsies versus resections, and unresectable stage III and first-line advanced/metastatic versus other disease stages. Based on these controlled trial data, pre-analytic variables of tissue specimens are a major driver of testing success; hence maintaining optimal conditions from sample collection to biomarker analysis, as well as improving tissue-sampling techniques is critical to increase testing success rates. TRIAL REGISTRATION NUMBERS NCT01802632, NCT02094261, NCT02151981, NCT02296125, NCT04035486, NCT02511106, NCT05120349, NCT03521154, NCT04351555.
Collapse
Affiliation(s)
| | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA, USA.
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute/Harvard Cancer Center, Boston, MA, USA.
| | - Jhanelle E Gray
- H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA.
| | - Myung-Ju Ahn
- Department of Hematology and Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Tom John
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia.
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan.
| | | | - Yuri Rukazenkov
- Oncology Research and Development, AstraZeneca, Cambridge, UK.
| | - Manana Javey
- Clinical Development Lead, Molecular Lab, Roche Diagnostics, Pleasanton, CA, USA.
| | - Helen Brown
- Precision Medicine & Biosamples, AstraZeneca, Cambridge, UK.
| | | |
Collapse
|
31
|
Mechahougui H, Gutmans J, Gouasmi R, Smekens L, Friedlaender A. BRAF Targeting Across Solid Tumors: Molecular Aspects and Clinical Applications. Int J Mol Sci 2025; 26:3757. [PMID: 40332392 PMCID: PMC12027668 DOI: 10.3390/ijms26083757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
BRAF mutations are critical drivers in cancers such as melanoma, colorectal cancer, and non-small-cell lung cancer. The most common mutation, BRAF V600E, is a key therapeutic target. Targeted treatments with BRAF and MEK inhibitors have significantly improved progression-free and overall survival in melanoma patients. However, in cancers like metastatic colorectal cancer, BRAF mutations are associated with poor outcomes due to aggressive disease behavior and resistance to conventional chemotherapy. Despite progress, resistance to BRAF/MEK inhibitors remains a major challenge, often driven by secondary mutations in the mitogen-activated protein kinase (MAPK) pathway, activation of alternative pathways such as phosphoinositide 3-kinases (PI3Ks)/protein kinase B (AKT), or changes in the tumor microenvironment. These challenges have motivated ongoing research into combining BRAF inhibitors with immunotherapies to enhance and prolong treatment effectiveness. Future research must also account for the role of the cancer's tissue of origin, as the biological context significantly influences response to targeted therapies, highlighting the need for a deeper understanding of tumor biology, micro-environment, and genetics.
Collapse
Affiliation(s)
- Hiba Mechahougui
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (J.G.); (L.S.)
| | - James Gutmans
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (J.G.); (L.S.)
| | - Roumaïssa Gouasmi
- Cancer Research Center of Lyon, CNRS UMR5286, Inserm U1052, University of Lyon, 69100 Lyon, France;
| | - Laure Smekens
- Oncology Department, Geneva University Hospital (HUG), 1205 Geneva, Switzerland; (J.G.); (L.S.)
| | | |
Collapse
|
32
|
Tajè R, Gallina FT, Caterino M, Forcella D, Patirelis A, Alessandrini G, Buglioni S, Cecere FL, Fusco F, Cappelli F, Melis E, Visca P, Cappuzzo F, Ambrogi V, Vidiri A. Molecular characterization of early-stage lung adenocarcinoma presenting as subsolid nodules in a real-life European cohort. BMC Cancer 2025; 25:647. [PMID: 40205411 PMCID: PMC11983824 DOI: 10.1186/s12885-025-13998-0] [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: 01/09/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
OBJECTIVES Subsolid nodules emerged as frequent radiological variants of lung adenocarcinoma. Radiological features including solid-component prevalence and larger tumour dimensions prompt tumoral invasiveness guiding prognosis and management. Thus, we aimed to clarify the molecular grounds that dictate these radiological appearances and clinical behaviour in a real-life European-cohort. Additionally, following the growing interest toward targeted-therapies in early-stage diseases, we aimed to present real-life epidemiological data of actionable mutations in these patients. METHODS In this retrospective single-centre study, targeted next-generation sequencing was performed continuatively in all the resected subsolid lung adenocarcinomas in the period between May 2016 and December 2023. Clinico-radiological data were collected. The genetic landscape of our real-life European subsolid adenocarcinoma population is defined. Common and actionable mutations (frequency > 5%) relation to key clinico-radiological features are evaluated. RESULTS Overall, 156 subsolid adenocarcinomas were analysed. KRAS-mutations, mostly KRAS p.G12C, were the most prevalent followed by EGFR, including 25% uncommon EGFR-mutations, TP53 and MET mutations. Amongst the clinico-radiological variables, KRAS-mutations and KRAS p.G12C-mutation were associated to smoking history (≥ 20 pack/years), aggressive histologic subtype and higher consolidation-to-tumor ratio (CTR). Moreover, KRAS-mutated nodules had faster tumour-doubling-time. Conversely, EGFR-mutations were associated to female sex and lower CTR. The latter not being confirmed in common EGFR-mutations. Additionally, in common EGFR-mutated nodules, aggressive histological components were rarer. CONCLUSION Our study presents the molecular profile of subsolid lung adenocarcinoma in a real-life European-cohort. KRAS-mutations were the most prevalent, and were related to smoking history, higher CTR and faster growth. Conversely, common EGFR-mutations were rarer than expected and unrelated to smoking history and radiological features.
Collapse
Affiliation(s)
- Riccardo Tajè
- Doctoral School of Microbiology, Immunology, Infectious Diseases and Transplants, MIMIT, University of Rome "Tor Vergata", Rome, Italy
- Thoracic Surgery Unit, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, 00144, Italy
| | - Filippo Tommaso Gallina
- Thoracic Surgery Unit, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, 00144, Italy.
- Tumor Immunology and Immunotherapy Unit, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy.
| | - Mauro Caterino
- Department of Radiology, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Daniele Forcella
- Thoracic Surgery Unit, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, 00144, Italy
| | - Alexandro Patirelis
- Doctoral School of Microbiology, Immunology, Infectious Diseases and Transplants, MIMIT, University of Rome "Tor Vergata", Rome, Italy
- Department of Thoracic Surgery, Tor Vergata University, Rome, Italy
| | - Gabriele Alessandrini
- Thoracic Surgery Unit, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, 00144, Italy
| | - Simonetta Buglioni
- Department of Pathology, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | | | - Francesca Fusco
- Medical Oncology 2, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Federico Cappelli
- Department of Radiology, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Enrico Melis
- Thoracic Surgery Unit, IRCCS "Regina Elena" National Cancer Institute, Via Elio Chianesi 53, Rome, 00144, Italy
| | - Paolo Visca
- Department of Pathology, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Federico Cappuzzo
- Medical Oncology 2, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Vincenzo Ambrogi
- Department of Thoracic Surgery, Tor Vergata University, Rome, Italy
| | - Antonello Vidiri
- Department of Radiology, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| |
Collapse
|
33
|
Faehling M, Fallscheer S, Schwenk B, Seifarth H, Sträter J, Lengerke C, Christopoulos P. Trends in Overall Survival in Lung Adenocarcinoma with EFGR Mutation, KRAS Mutation, or No Mutation. Cancers (Basel) 2025; 17:1237. [PMID: 40227775 PMCID: PMC11988053 DOI: 10.3390/cancers17071237] [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: 03/10/2025] [Revised: 03/30/2025] [Accepted: 04/02/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Treatment of lung adenocarcinoma has changed and now includes checkpoint inhibitors (CPIs) or, in the case of an EGFR mutation, third-generation EGFR TKI osimertinib. Few data compare the long-term overall survival (OS) of current and historic subgroups. METHODS This real-world analysis (KOMPASS study) included stage IV lung-adenocarcinoma patients with either EGFR, KRAS, or no mutation. Patients were assigned to the "current" EGFR, KRAS, or no-mutation cohort if they had mutation testing using NGS (n = 199; median date of diagnosis 2021). If they had an EGFR PCR test only, they were assigned to the "historic" EGFR or no-mutation cohort (n = 127; median date of diagnosis 2014). RESULTS Both the current and the historic EGFR cohorts had significantly longer OS than the respective no-mutation cohorts (HR 0.58 and 0.60, respectively). The current no-mutation and EGFR cohorts had a strong trend to longer OS than the respective historic cohorts. In the no-mutation cohorts, the improvement was due to an increase in long-term survivors (HR 0.71), whereas in the EGFR mutation cohorts, the median OS was improved without long-term survivors (HR 0.70). The KRAS cohort showed OS like the no-mutation cohort, with a plateau of long-term survivors around 20%. CONCLUSIONS A comparison of our data with that of the phase III trials KEYNOTE-189 and FLAURA suggests that the improved outcomes are due to the use of CPIs or osimertinib. The clinical trial results are well translated into real-world clinical practice with comparable OS. KRAS patients benefit from CPI treatment like no-mutation patients.
Collapse
Affiliation(s)
- Martin Faehling
- Clinic of Cardiology and Pneumology, Esslingen Hospital, 73730 Esslingen, Germany
| | - Sabine Fallscheer
- Clinic of Cardiology and Pneumology, Esslingen Hospital, 73730 Esslingen, Germany
| | - Birgit Schwenk
- Clinic of Cardiology and Pneumology, Esslingen Hospital, 73730 Esslingen, Germany
| | - Harald Seifarth
- Clinic of Radiology, University of Münster, 48143 Münster, Germany
- Clinic of Radiology, Esslingen Hospital, 73730 Esslingen, Germany
| | - Jörn Sträter
- Institute of Pathology, 73730 Esslingen, Germany
| | - Claudia Lengerke
- University Hospital Tübingen, Department for Internal Medicine II, University of Tübingen, 72076 Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, a Partnership Between DKFZ and University Hospital Tübingen, 72076 Tübingen, Germany
| | - Petros Christopoulos
- Department of Medical Oncology, Thorax Clinic, 69126 Heidelberg, Germany
- National Center for Tumor Diseases, Heidelberg University Hospital, 69126 Heidelberg, Germany
| |
Collapse
|
34
|
Sayin SI, Eklund EA, Ali KX, Dzanan JJ, Xylander M, Dankis M, Lindahl P, Sayin VI, Hallqvist A, Wiel C. Distinct metastatic organotropism shapes prognosis in lung adenocarcinoma with brain metastasis. Front Oncol 2025; 15:1569517. [PMID: 40291914 PMCID: PMC12031661 DOI: 10.3389/fonc.2025.1569517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/18/2025] [Indexed: 04/30/2025] Open
Abstract
Background Metastatic organotropism in lung cancer significantly influences prognosis, yet current treatment and clinical management guidelines are largely generalized for metastatic disease, regardless of organ site involvement. Notably, up to 30% of non-small cell lung cancer (NSCLC) patients present with brain metastases (BM) at diagnosis, underscoring the need for a more nuanced understanding of metastatic patterns. However, real-world clinical data on metastatic organotropism in well-characterized patient cohorts remain surprisingly scarce. Here, we evaluate patterns of metastasis, clinical characteristics and survival outcomes in patients with lung adenocarcinoma (LUAD), the major histological NSCLC subtype. Methods We performed a multi-center retrospective study including 913 stage IV LUAD patients, diagnosed and molecularly assessed in western Sweden between 2016-2021. Our primary study outcome was the distribution of specific metastatic sites and its impact on Overall Survival (OS). Results Out of 913 stage IV LUAD patients, 23.4% had BM. These patients exhibited markedly different metastatic patterns compared to those without BM, and median survival was significantly shorter (6 months) than those without BM (7.8 months) (p = 0.021). In addition, more than one metastatic tumor in the brain coincided with worse OS, compared to those with no, or with only one metastatic tumor in the brain. Importantly, OS was also influenced by metastasis in specific extracranial organs, like the pleura and lungs. Conclusions Our study highlights the distinct metastatic patterns and survival outcomes associated with BM in stage IV LUAD. These findings emphasize the need for site-specific approaches in managing metastatic disease due to BM's impact on survival.
Collapse
Affiliation(s)
- Sama I. Sayin
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
| | - Ella A. Eklund
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Kevin X. Ali
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jozefina J. Dzanan
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Moe Xylander
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
| | - Martin Dankis
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Per Lindahl
- Department of Molecular and Clinical Medicine, Institute of Medicine University of Gothenburg, Gothenburg, Sweden
- Department of Biochemistry, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Volkan I. Sayin
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Andreas Hallqvist
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Oncology, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Clotilde Wiel
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
35
|
Torasawa M, Yoshida T, Shiraishi K, Yagishita S, Ono H, Uehara Y, Miyakoshi J, Tateishi A, Igawa YS, Higashiyama RI, Mochizuki A, Masuda K, Matsumoto Y, Shinno Y, Okuma Y, Goto Y, Horinouchi H, Hamamoto R, Yamamoto N, Watanabe SI, Yatabe Y, Takahashi K, Kohno T, Ohe Y. Implications of EGFR expression on EGFR signaling dependency and adaptive immunity against EGFR-mutated lung adenocarcinoma. Lung Cancer 2025; 202:108494. [PMID: 40088580 DOI: 10.1016/j.lungcan.2025.108494] [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: 11/03/2024] [Revised: 02/09/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND In EGFR-mutated lung adenocarcinoma (EGFRm LUAD), EGFR mutations do not necessarily result in increased EGFR expression (EGFR-exp), which differs among patients. However, the factors influencing EGFR-exp and the impact of EGFR-exp on tumor characteristics in patients with EGFRm LUAD remain unclear. PATIENTS AND METHODS Whole-exome and RNA sequencing were performed for patients with early- and advanced-stage EGFRm LUAD. The patients were classified into low or high EGFR-exp groups based on the median transcripts per million. We retrospectively examined the association between EGFR-exp, genomic characteristics, downstream EGFR signaling activity, tumor microenvironment (TME) status, and clinical outcomes. RESULTS This study included 450 and 45 patients in the early- and advanced-stage cohorts, respectively. In both cohorts, the EGFR-exp low group exhibited a lower incidence of TP53 co-mutations and EGFR amplification and a higher incidence of EGFR subclonal mutations than the EGFR-exp high group. Furthermore, downstream EGFR signaling pathways, such as the MAPK signaling, were less activated in the EGFR-exp low group. However, this group showed significantly enriched adaptive immune response pathways (Q < 0.0001) and an immune-inflamed TME. Additionally, a low EGFR-exp was a significantly favorable factor for postoperative relapse (odds ratio [OR], 0.6; P = 0.04). However, in the advanced-stage cohort, a low EGFR-exp was a significant risk factor for non-responders to osimertinib (OR, 17.5; P = 0.03). CONCLUSIONS In EGFRm LUAD, significant associations were observed between EGFR-exp levels and both EGFR signaling pathways and adaptive immune status, which in turn influence clinical outcomes. This large-scale multi-omics analysis highlights the heterogeneity among patients with EGFRm LUAD and emphasizes the need to assess EGFR-exp levels alongside mutation status for optimal treatment strategies in EGFRm LUAD.
Collapse
Affiliation(s)
- Masahiro Torasawa
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan; Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan; Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Tatsuya Yoshida
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan; Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Shigehiro Yagishita
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Hanako Ono
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Yuji Uehara
- Division of Cancer Evolution, National Cancer Center Research Institute, Tokyo, Japan; Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Jun Miyakoshi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Akiko Tateishi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | | | | | - Akifumi Mochizuki
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Ken Masuda
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuji Matsumoto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuki Shinno
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yusuke Okuma
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yasushi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ryuji Hamamoto
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Noboru Yamamoto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan; Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Shun-Ichi Watanabe
- Department of Thoracic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
36
|
Shi Y, Guo Y, Li X, Wu L, Chen Z, Yang S, Bi M, Zhao Y, Yao W, Yu H, Wang K, Zhao W, Sun M, Zhang L, He Z, Lin Y, Shi J, Zhu B, Wang L, Pan Y, Shi H, Sun S, Wen M, Zhou R, Guo S, Han Z, Yi T, Zhang H, Cang S, Yu Z, Zhong D, Cui J, Fang J, Gao J, Li M, Ma R, Jiang M, Qin J, Shu Y, Ye F, Hu S, Li W, Lu H, Yang M, Yi S, Zhang Y, Fan Y, Ji H, Liu Z, Wang H, Zhou X, Zhang D, Peng J, Shen H, Gao F, Wang T, Zhou A. Rezivertinib versus gefitinib as first-line therapy for patients with EGFR-mutated locally advanced or metastatic non-small-cell lung cancer (REZOR): a multicentre, double-blind, randomised, phase 3 study. THE LANCET. RESPIRATORY MEDICINE 2025; 13:327-337. [PMID: 39914443 DOI: 10.1016/s2213-2600(24)00417-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 03/17/2025]
Abstract
BACKGROUND This study aimed to compare the efficacy and safety of rezivertinib (BPI-7711) and gefitinib as first-line therapies in patients with EGFR-mutated locally advanced or metastatic non-small-cell lung cancer (NSCLC). METHODS This multicentre, double-blind, randomised, phase 3 study (REZOR) included eligible patients from 50 hospitals across China. Those who had been histologically or cytologically confirmed as having NSCLC with EGFR exon 19 deletion or exon 21 Leu858Arg mutation by central laboratory were randomly assigned (1:1) to receive once daily either rezivertinib 180 mg or gefitinib 250 mg, until unacceptable toxicity occurred, disease progression, or other treatment discontinuation criteria were met. Each cycle lasted for 21 days. The primary endpoint was progression-free survival evaluated by masked independent central review (MICR) in the intention-to-treat set. This trial is registered with ClinicalTrials.gov, NCT03866499 and follow-up is ongoing. FINDINGS Between July 15, 2019, and Feb 14, 2022, 695 patients were screened. Among them, 369 eligible patients were randomly assigned to receive either rezivertinib 180 mg/day plus placebo (n=184) or gefitinib 250 mg/day plus placebo (n=185) in a 1:1 ratio; all of eligible participants were included in the intention-to-treat set. Median MICR-assessed progression-free survival was 19·3 months (95% CI 13·8-22·1) in the rezivertinib group and 9·6 months (8·4-11·3) in the gefitinib group (hazard ratio [HR] 0·48, 95% CI 0·36-0·63; p<0·0001) and the prespecified subgroup efficacy analysis showed consistent results. Median duration of exposure was 16·0 months (95% CI 0·0-29·7) in the rezivertinib group and 11·0 months (0·0-28·9) in the gefitinib group. Grade 3 or higher treatment-emergent adverse events (82 [45%] of 184 in the rezivertinib group; 80 [43%] of 185 in the gefitinib group) and treatment-related adverse events (TRAEs; 43 [23%] of 184 in the rezivertinib group; 43 [23%] of 185 in the gefitinib group) were similar in both groups. One patient died from a TRAE in the rezivertinib group, due to pneumonia and interstitial lung disease. INTERPRETATION Our findings suggested that rezivertinib is a potential choice for patients with EGFR-mutated locally advanced or metastatic NSCLC as first-line therapy, owing to the superior overall efficacy and subgroup progression-free survival compared with gefitinib in targeted patients. No new safety signals were identified. FUNDING Beta Pharma (Shanghai) and the China National Science and Technology Major Project for Key New Drug Development.
Collapse
Affiliation(s)
- Yuankai Shi
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Yanzhen Guo
- Department of Medical Oncology, The First Affiliated Hospital of Henan University of Science & Technology, Luoyang, China
| | - Xingya Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin Wu
- Department of Thoracic Medical Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhaohong Chen
- Department of Oncology, People's Hospital of Deyang City, Deyang, China
| | - Sheng Yang
- Department of Medical Oncology, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Minghong Bi
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Yanqiu Zhao
- Respiratory Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Wenxiu Yao
- Department of Medical Oncology, Sichuan Cancer Hospital-Cancer Hospital Affiliated to University of Electronic Science and Technology of China, Chengdu, China
| | - Huiqing Yu
- Department of Geriatric Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Ke Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Wenhua Zhao
- Department of Internal Medicine for Lung Cancer, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Meili Sun
- Department of Oncology, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Liangming Zhang
- Department of Medical Oncology, Yantai Yuhuangding Hospital, Yantai, China
| | - Zhiyong He
- Thoracic Medical Oncology, Fujian Cancer Hospital, Fuzhou, China
| | - Yingcheng Lin
- Department of Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Jianhua Shi
- Department of Medical Oncology, Linyi Cancer Hospital, Linyi, China
| | - Bo Zhu
- Department of Oncology, Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Lijun Wang
- Cancer Center, The Second Affiliated Hospital of Xingtai Medical College, Xingtai, China
| | - Yueyin Pan
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huaqiu Shi
- Department of Medical Oncology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Shenghua Sun
- Department of Respiratory Medicine, Third Xiangya Hospital of Central South University, Changsha, China
| | - Meiling Wen
- Department of Medical Oncology, The First Affiliated Hospital of the University of South China, Hengyang, China
| | - Rui Zhou
- Department of Respiratory Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuliang Guo
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhigang Han
- Pulmonary Cancer Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
| | - Tienan Yi
- Department of Medical Oncology, Xiangyang Central Hospital, Xiangyang, China
| | - Hua Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Shundong Cang
- Department of Oncology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhuang Yu
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - DianSheng Zhong
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiuwei Cui
- Oncology Center, Oncology Department, The First Hospital of Jilin University, Changchun, China
| | - Jian Fang
- Department of Thoracic Oncology, Beijing Cancer Hospital, Beijing, China
| | - Jinghua Gao
- Department of Medical Oncology, Cangzhou Central Hospital, Cangzhou, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rui Ma
- Department of Thoracic Oncology, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Mingyan Jiang
- Department of Respiratory and Critical Care Medicine, Xiangtan Central Hospital, Xiangtan, China
| | - Jianwen Qin
- Respiratory and Critical Care Department, Tianjin Chest Hospital, Tianjin, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing, China
| | - Feng Ye
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Sheng Hu
- Department of Thoracic Oncology, Hubei Cancer Hospital, Wuhan, China
| | - Wen Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Lu
- Department of Oncology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Minglei Yang
- Department of Thoracic Surgery, Ningbo No.2 Hospital, Ningbo, China
| | - Shanyong Yi
- Department of Medical Oncology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Yan Zhang
- Department of Medical Oncology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Yun Fan
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Hongbo Ji
- Department of Medical Oncology in Section One, Chifeng Municipal Hospital, Chifeng, China
| | - Zheng Liu
- Department of Oncology, Handan Central Hospital, Handan, China
| | - Haitao Wang
- Department of Medical Oncology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xiangdong Zhou
- Department of Respiratory and Critical Care Medicine, The first affiliated Hospital of the Army Medical University of Chinese People's Liberation Army, Chongqing, China
| | - Don Zhang
- Department of Drug Discovery, Beta Pharma, Princeton, NJ, USA
| | - Jirong Peng
- Department of Drug Discovery, Beta Pharma, Princeton, NJ, USA
| | - Haijiao Shen
- Department of Clinical Development, Beta Pharma (Shanghai), Shanghai, China
| | - Feng Gao
- Department of Clinical Development, Beta Pharma (Shanghai), Shanghai, China
| | - Tingting Wang
- Department of Clinical Development, Beta Pharma (Shanghai), Shanghai, China
| | - Anqi Zhou
- Department of Clinical Development, Beta Pharma (Shanghai), Shanghai, China
| |
Collapse
|
37
|
Sands J, Ahn MJ, Lisberg A, Cho BC, Blumenschein G, Shum E, Pons Tostivint E, Goto Y, Yoh K, Heist R, Shimizu J, Lee JS, Baas P, Planchard D, Pérol M, Felip E, Su WC, Zebger-Gong H, Lan L, Liu C, Howarth P, Chiaverelli R, Paz-Ares L. Datopotamab Deruxtecan in Advanced or Metastatic Non-Small Cell Lung Cancer With Actionable Genomic Alterations: Results From the Phase II TROPION-Lung05 Study. J Clin Oncol 2025; 43:1254-1265. [PMID: 39761483 PMCID: PMC11949215 DOI: 10.1200/jco-24-01349] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/05/2024] [Accepted: 11/14/2024] [Indexed: 03/29/2025] Open
Abstract
PURPOSE Datopotamab deruxtecan (Dato-DXd) is a trophoblast cell-surface antigen-2-directed antibody-drug conjugate with a highly potent topoisomerase I inhibitor payload. The TROPION-Lung05 phase II trial (ClinicalTrials.gov identifier: NCT04484142) evaluated the safety and clinical activity of Dato-DXd in patients with advanced/metastatic non-small cell lung cancer (NSCLC) with actionable genomic alterations progressing on or after targeted therapy and platinum-based chemotherapy. PATIENTS AND METHODS Patients received Dato-DXd 6 mg/kg once every 3 weeks. The primary end point was objective response rate (ORR) by blinded independent central review. Secondary end points included duration of response (DOR), safety, tolerability, and survival. RESULTS Among 137 patients who received at least 1 dose of Dato-DXd, 71.5% received at least three lines of prior therapies for advanced/metastatic disease. Overall, 56.9% had EGFR mutations and 24.8% had ALK rearrangements. Median treatment duration was 4.4 months (range, 0.7-20.6). The confirmed ORR was 35.8% (95% CI, 27.8 to 44.4) overall, and 43.6% (95% CI, 32.4 to 55.3) and 23.5% (95% CI, 10.7 to 41.2) in those with EGFR mutations and ALK rearrangements, respectively. The median DOR was 7.0 months (95% CI, 4.2 to 9.8), and the overall disease control rate was 78.8% (95% CI, 71.0 to 85.3). Grade ≥3 treatment-related adverse events (TRAEs) occurred in 28.5% of patients. The most common TRAE was stomatitis (preferred term; any grade: 56.2%; grade ≥3: 9.5%). Five (3.6%) patients experienced adjudicated treatment-related interstitial lung disease/pneumonitis, with 1 (0.7%) grade 5 event. CONCLUSION Encouraging and durable antitumor activity was observed with Dato-DXd in this heavily pretreated advanced/metastatic NSCLC population with actionable genomic alterations. The rate of treatment-related grade ≥3 toxicities was comparable with previous observations, and no new safety signals were observed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Kiyotaka Yoh
- National Cancer Center Hospital East, Kashiwa, Japan
| | - Rebecca Heist
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Harvard University, Boston, MA
| | | | - Jong-Seok Lee
- Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Paul Baas
- Dana-Farber Cancer Institute, Boston, MA
| | | | | | | | - Wu-Chou Su
- Dana-Farber Cancer Institute, Boston, MA
| | | | - Lan Lan
- Daiichi Sankyo, Inc, Basking Ridge, NJ
| | | | | | | | | |
Collapse
|
38
|
Trojnar A, Domagała-Kulawik J. Current insights into the clinico-pathologic characteristics of lung cancer in women. Expert Rev Respir Med 2025; 19:301-312. [PMID: 40040469 DOI: 10.1080/17476348.2025.2475974] [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: 11/12/2024] [Revised: 01/14/2025] [Accepted: 03/03/2025] [Indexed: 03/06/2025]
Abstract
INTRODUCTION Lung cancer is responsible for premature cancer deaths in women and is the first cause of cancer deaths in women in many countries. The problem of lung cancer in women seems to be underestimated in many aspects, including low participation in clinical trials and screening tests. AREAS COVERED Current research progress has contributed to a better understanding of the issue and makes it possible to describe the problem in a new light. In our paper, the problem of lung cancer in women was discussed in a broad aspect, taking into account women's health, the harmful effects of smoking and the current diagnostic and treatment process. The results of treatment also differ in relation to sex. All these aspects of the diversity of women's lung cancer were presented on the basis of newest and most comprehensive literature. EXPERT OPINION Lung cancer in women is and will remain an important health problem worldwide, which is justified by epidemiological data, basic research and treatment results.
Collapse
Affiliation(s)
- Anna Trojnar
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
39
|
Heersche N, Lanser DAC, Muntinghe-Wagenaar MB, Mohmaed Ali MI, Ulas EB, Trooster TMA, de Jonge E, Oomen-de Hoop E, Paats MS, Bahce I, Croes S, Hendriks LEL, van der Wekken AJ, Dingemans AMC, Huitema ADR, van Schaik RHN, Mathijssen RHJ, Veerman GDM. Sex and Common Germline Variants Affect the Toxicity Profile and Pharmacokinetics of Alectinib: A Nationwide Cohort Study in Patients With ALK-Positive NSCLC. J Thorac Oncol 2025; 20:475-486. [PMID: 39617342 DOI: 10.1016/j.jtho.2024.11.025] [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: 06/25/2024] [Revised: 10/30/2024] [Accepted: 11/25/2024] [Indexed: 12/22/2024]
Abstract
INTRODUCTION Alectinib, a small-molecule kinase inhibitor, is used as first-line treatment for ALK-positive (ALK+) NSCLC. Albeit generally well-tolerated, a considerable subset of patients requires dose adjustments due to drug-related toxicity. Single-nucleotide polymorphisms in genes related to the metabolism of alectinib may upfront identify patients at risk for toxicity. METHODS In this multicenter observational cohort study in patients with advanced ALK+ NSCLC receiving alectinib treatment, we investigated the association between toxicity, pharmacokinetics, and key genetic variants in ABCB1, CYP3A4, PPAR-α, POR, and CYP3A5. Data on demographics, adverse events, and alectinib trough levels were collected from five hospitals. RESULTS Among 215 patients, 47% experienced severe toxicity. Women experienced more severe toxicity (female versus male: 56% versus 34%; p = 0.001) and had +35% higher alectinib trough levels (p < 0.001). Homozygous carriers of the PPAR-α 209G>A variant exhibited a higher incidence of grade greater than or equal to 3 toxicity (38%) compared with patients who carried at least one wild-type allele (11%) (p = 0.004). This remained significant after Bonferroni correction. Patients who experienced severe toxicity had +18.5% (95% confidence interval: 2.9%-36.6%; p = 0.019) higher trough levels. CONCLUSIONS Female patients encounter more severe toxicity due to higher alectinib exposure, which warrants further exploration. PPAR-α 209G>A significantly increased relevant alectinib-induced toxicity, most likely due to an increase in alectinib exposure. Pretreatment testing for genetic variants with a subsequent dose reduction could provide a viable approach to reduce alectinib-related toxicity.
Collapse
Affiliation(s)
- Niels Heersche
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Daan A C Lanser
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M Benthe Muntinghe-Wagenaar
- Department of Pulmonary Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ma Ida Mohmaed Ali
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ezgi B Ulas
- Department of Pulmonary Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Tessa M A Trooster
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Evert de Jonge
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Esther Oomen-de Hoop
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Marthe S Paats
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Idris Bahce
- Department of Pulmonary Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Sander Croes
- Department of Clinical Pharmacy & Toxicology, Maastricht University Medical Center, CARIM - School for Cardiovascular Disease, Maastricht, The Netherlands
| | - Lizza E L Hendriks
- Department of Pulmonary Medicine, Maastricht University Medical Center, GROW - School for Oncology and Reproduction, Maastricht, The Netherlands
| | - Anthonie J van der Wekken
- Department of Pulmonary Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anne-Marie C Dingemans
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute, Amsterdam, The Netherlands; Department of Pharmacology, Princess Máxima Center for Paediatric Oncology, Utrecht, The Netherlands; Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ron H N van Schaik
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - G D Marijn Veerman
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands; Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
40
|
Nagasaka M, Roy UB, Berk A, Liu G, Nadler E, Abrahami D. The value of real-world evidence in supporting targeted therapies for patients with rare oncogenic drivers in mNSCLC. Future Oncol 2025; 21:1005-1011. [PMID: 40084656 PMCID: PMC11988244 DOI: 10.1080/14796694.2025.2475728] [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: 12/19/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
With the ongoing discovery of various oncogenic driver mutations in metastatic non-small cell lung cancer (mNSCLC), a precision medicine approach has emerged, characterized by targeted therapies for select patient populations. Randomized controlled trials (RCT) remain the gold standard for evaluating efficacy and safety of such therapies; however, RCTs evaluating treatments for rare oncogenic drivers still face limitations, given small populations, potentially long-time horizon for outcome events to occur, and underrepresentation of certain subgroups. For these targeted therapies, the complementary nature between real-world evidence (RWE) and RCT may expand the totality of evidence available, to better inform treatment decision-making. In particular, treatments for rare oncogenic drivers can benefit from RWE that provides additional, generalizable clinical insights for subgroups underrepresented or ineligible for RCT, or confirms outcomes observed in RCT. As a discipline, RWE has seen significant advances in methodology and healthcare stakeholder acceptability, with potential for even greater innovation, and presents a valuable opportunity to support decision-making around access and use of targeted therapies for rare oncogenic drivers in mNSCLC.
Collapse
Affiliation(s)
- Misako Nagasaka
- Division of Hematology and Oncology, Department of Medicine, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA, USA
- Division of Neurology, Department of Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Upal Basu Roy
- Translational Science Research Program, LUNGevity Foundation, Chicago, IL, USA
| | | | - Geoffrey Liu
- Medical Oncology and Hematology, Princess Margaret Cancer Centre, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Eric Nadler
- Charles Sammons Cancer Center, Baylor University Medical Center, Dallas, TX, USA
| | - Devin Abrahami
- HTA Value and Evidence, Oncology, Pfizer Inc, New York, NY, USA
| |
Collapse
|
41
|
Chung C, Umoru G. Prognostic and predictive biomarkers with therapeutic targets in nonsmall-cell lung cancer: A 2023 update on current development, evidence, and recommendation. J Oncol Pharm Pract 2025; 31:438-461. [PMID: 38576390 DOI: 10.1177/10781552241242684] [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: 04/06/2024]
Abstract
BackgroundSince the publication of the original work in 2014, significant progress has been made in the characterization of genomic alterations that drive oncogenic addiction of nonsmall cell lung cancer (NSCLC) and how the immune system can leverage non-oncogenic pathways to modulate therapeutic outcomes. This update evaluates and validates the recent and emerging data for prognostic and predictive biomarkers with therapeutic targets in NSCLC.Data sourcesWe performed a literature search from January 2015 to October 2023 using the keywords non-small cell lung cancer, clinical practice guidelines, gene mutations, genomic assay, immune cancer therapy, circulating tumor DNA, predictive and prognostic biomarkers, and targeted therapies.Study selection and data extractionWe identified, reviewed, and evaluated relevant clinical trials, meta-analyses, seminal articles, and published clinical practice guidelines in the English language.Data synthesisRegulatory-approved targeted therapies include those somatic gene alterations of EGFR ("classic" mutations, exon 20 insertion, and rare EGFR mutations), ALK, ROS1, BRAF V600, RET, MET, NTRK, HER2, and KRAS G12C. Data for immunotherapy and circulating tumor DNA in next-generation sequencing are considered emerging, whereas the predictive role for PIK3CA gene mutation is insufficient.ConclusionsAdvances in sequencing and other genomic technologies have led to identifying novel oncogenic drivers, novel resistance mechanisms, and co-occurring mutations that characterize NSCLC, creating further therapeutic opportunities. The benefits associated with immunotherapy in the perioperative setting hold initial promise, with their long-term results awaiting.
Collapse
Affiliation(s)
- Clement Chung
- Department of Pharmacy, Houston Methodist West Hospital, Houston, TX, USA
| | - Godsfavour Umoru
- Department of Pharmacy, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
42
|
Cai JQ, Wang YM, Lin X, Xie M, Zhang G, Wei XX, Sun H. Cardiovascular toxicity of anaplastic lymphoma kinase inhibitors for patients with non-small cell lung cancer: a network meta-analysis. Future Oncol 2025; 21:1125-1135. [PMID: 39400073 DOI: 10.1080/14796694.2024.2370239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 06/17/2024] [Indexed: 10/15/2024] Open
Abstract
Aim: We conducted network meta-analysis to assess cardiovascular toxicity of anaplastic lymphoma kinase-tyrosine kinase inhibitors (ALK-TKIs).Materials & methods: Eleven articles involving 2855 patients and six interventions including crizotinib, alectinib, ceritinib, lorlatinib, brigatinib and chemotherapy were analyzed.Results: No significant difference was observed in overall cardiovascular risk among ALK-TKIs. Subgroup analysis showed that for cardiac toxicity, crizotinib and alectinib were more likely to cause myocardial rhythm abnormalities. Crizotinib and ceritinib had a higher risk of Q-T prolongation than chemotherapy. For vascular toxicity, crizotinib and ceritinib had a higher risk of thrombotic events than brigatinib. Crizotinib and lorlatinib were more likely to cause blood pressure abnormalities.Conclusion: Clinicians should carefully monitoring cardiovascular events when ALK-TKIs used in NSCLCs patients with baseline cardiovascular diseases.
Collapse
Affiliation(s)
- Jia Qin Cai
- Department of Pharmacy, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Yi Ming Wang
- School of Pharmacy, Fujian University of Traditional Chinese Medicine, Department of Pharmacy, Fuzhou, Fujian, China
| | - Xinmiao Lin
- School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Mumu Xie
- Department of Pharmacy, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Guifeng Zhang
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Xiao Xia Wei
- Department of Pharmacy, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Hong Sun
- Department of Pharmacy, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| |
Collapse
|
43
|
Soo RA, Vervita K, Früh M, Cho BC, Majem M, Rodriguez Abreu D, Ribi K, Callejo A, Moran T, Domine Gomez M, Provencio M, Addeo A, Han JY, Ortega Granados AL, Reck M, Blasco A, Garcia Campelo R, Sala González MA, Britschgi C, Roschitzki-Voser H, Ruepp B, Gasca-Ruchti A, Haberecker M, Dafni U, Peters S, Stahel RA. A randomised non-comparative phase II study of atezolizumab, bevacizumab and chemotherapy in EGFR-mutant NSCLC with acquired resistance - The ETOP 15-19 ABC-lung trial. Lung Cancer 2025; 202:108454. [PMID: 40023017 DOI: 10.1016/j.lungcan.2025.108454] [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: 12/06/2024] [Revised: 01/29/2025] [Accepted: 02/16/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND ABC-lung explores the potential effect of combining atezolizumab and bevacizumab with either carboplatin/paclitaxel (ABCPac) or pemetrexed (ABPem) in patients with EGFR-mutant NSCLC, resistant to tyrosine kinase inhibitors (TKIs). METHODS ABC-lung is a 1:1 randomised, non-comparative, phase II trial, stratified by prior treatment with a third-generation EGFR TKI, evaluating atezolizumab (1200 mg, Q3W) and bevacizumab (15mg/kg, Q3W) with either 4-6 cycles of carboplatin (AUC5, Q3W) and paclitaxel (175-200mg/m2, Q3W) or pemetrexed (500 mg/m2, Q3W) until progression (PD). The study aimed to improve the 1-year progression-free survival (PFS) rate from 18% to 37%, assessed per RECISTv1.1, separately in each arm. To reject the null hypothesis, at least 14 of 45 evaluable patients in each arm needed to be progression-free at 1-year (power 83%, 1-sided a=0.023). Secondary endpoints included overall survival (OS), objective response rate (ORR), PFS, quality of life (QoL) and adverse events (AEs). RESULTS Between 09/2020 and 09/2022, 95 patients were randomized (ABCPac:45; ABPem:50) with median follow-up time of 19 months. From the evaluable patients, 9 in ABCPac and 11 in ABPem arms reached 1-year without progression, lower than the success criterion of 14patients. Median PFS was 6.4 months in ABCPac and 7.6 months in the ABPem arms, while median OS was 15.4 months and 15.6 months, respectively. Grade ≥3 treatment-related AEs were experienced by 50% and 42% of patients in ABCPac and ABPem arms, respectively, while no grade 5 AEs were recorded. CONCLUSIONS The observed 1-year PFS rate with atezolizumab, bevacizumab in combination with either carboplatin-paclitaxel or pemetrexed was below the aspired rate of 37% in both arms. The safety is consistent with the known toxicity profiles. Clinical trial identification: NCT04245085.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- Bevacizumab/administration & dosage
- Bevacizumab/therapeutic use
- Lung Neoplasms/drug therapy
- Lung Neoplasms/mortality
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Female
- Male
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Middle Aged
- Aged
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Drug Resistance, Neoplasm
- ErbB Receptors/genetics
- Mutation
- Adult
- Paclitaxel/administration & dosage
- Carboplatin/administration & dosage
- Pemetrexed/administration & dosage
- Aged, 80 and over
Collapse
Affiliation(s)
- R A Soo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - K Vervita
- ETOP Statistical Office, FSF-H - Frontier Science Foundation Hellas, Athens, Greece
| | - M Früh
- Klinik für Medizinische Onkologie und Hämatologie, Kantonsspital St. Gallen, St. Gallen, Switzerland; Department of Medical Oncology, University of Bern, Bern, Switzerland
| | - B C Cho
- Medical Oncology Department, Yonsei University, Seoul, Republic of Korea
| | - M Majem
- Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Spanish Lung Cancer Group (GECP), Barcelona, Spain
| | - D Rodriguez Abreu
- Spanish Lung Cancer Group (GECP), Barcelona, Spain; Medical Oncology Department, Hospital Universitario Insular de Gran Canaria - Complejo Hospitalario Materno-Insular, Las Palmas De Gran Canaria, Spain
| | - K Ribi
- Coordinating Center, ETOP IBCSG Partners Foundation, Bern, Switzerland
| | - A Callejo
- Spanish Lung Cancer Group (GECP), Barcelona, Spain; Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | - T Moran
- Spanish Lung Cancer Group (GECP), Barcelona, Spain; Medical Oncology Dept, ICO - Institut Català d'Oncologia Badalona (Hospital Universitario Germans Trias i Pujol), Badalona, Spain
| | - M Domine Gomez
- Spanish Lung Cancer Group (GECP), Barcelona, Spain; Oncology Dept., Hospital Universitario Fundacion Jimenez Diaz, IIS-FJD, Madrid, Spain
| | - M Provencio
- Spanish Lung Cancer Group (GECP), Barcelona, Spain; Universidad Autónoma de Madrid, University Hospital Puerta de Hierro Majadahonda, Majadahonda, Spain
| | - A Addeo
- Oncology Dept., HUG - Hopitaux Universitaires de Geneve, Geneva, Switzerland
| | - J Y Han
- Center for Lung Cancer, National Cancer Center, Goyang, Republic of Korea
| | - A L Ortega Granados
- Spanish Lung Cancer Group (GECP), Barcelona, Spain; Medical Oncology Department, Hospital Universitario de Jaén, Jaén, Spain
| | - M Reck
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), LungenClinic, Grosshansdorf, Germany
| | - A Blasco
- Spanish Lung Cancer Group (GECP), Barcelona, Spain; Hospital General de Valencia, Spain
| | - R Garcia Campelo
- Spanish Lung Cancer Group (GECP), Barcelona, Spain; Hospital Teresa Herrera, La Coruña, Spain
| | - M A Sala González
- Spanish Lung Cancer Group (GECP), Barcelona, Spain; Medical Oncology Department, Hospital Universitario Basurto, Bilbao, Spain
| | - C Britschgi
- Medical Oncology and Haematology, Universitätsspital Zürich (USZ), Zürich, Switzerland
| | | | - B Ruepp
- Coordinating Center, ETOP IBCSG Partners Foundation, Bern, Switzerland
| | - A Gasca-Ruchti
- Coordinating Center, ETOP IBCSG Partners Foundation, Bern, Switzerland
| | - M Haberecker
- Department of Pathology and Molecular Pathology, USZ - University Hospital Zurich, Zurich, Switzerland
| | - U Dafni
- Department of Medical Oncology, University of Bern, Bern, Switzerland; National and Kapodistrian University of Athens, Athens, Greece
| | - S Peters
- Oncology Dept., CHUV - Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - R A Stahel
- Coordinating Center, ETOP IBCSG Partners Foundation, Bern, Switzerland.
| |
Collapse
|
44
|
Jin S, He Y, Feng C, Yuan J, Guo Y, Guo Z, Wang X. Cellular Discrepancy of Platinum Complexes in Interfering with Mitochondrial DNA. ACS CENTRAL SCIENCE 2025; 11:393-403. [PMID: 40161961 PMCID: PMC11950849 DOI: 10.1021/acscentsci.4c01941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 04/02/2025]
Abstract
Mitochondria are associated with cellular energy metabolism, proliferation, and mode of death. Damage to mitochondrial DNA (mtDNA) greatly affects mitochondrial function by interfering with energy production and the signaling pathway. Monofunctional trinuclear platinum complex MTPC demonstrates different actions on the mtDNA of cancerous and normal cells. It severely impairs the integrity and function of mitochondria in the human lung cancer A549 cells, such as dissipating mitochondrial membrane potential, decreasing the copy number of mtDNA, interfering in nucleoid proteins and polymerase gamma gene, reducing adenosine triphosphate (ATP), and inducing mitophagy, whereas it barely affects the mtDNA of the human kidney 2 (HK-2) cells. Moreover, MTPC promotes the release of mtDNA into the cytosol and stimulates the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway, thus showing the potential to trigger antitumor immunity. MTPC displays significant cytotoxicity against A549 cells, while it exhibits weak toxicity toward HK-2 cells, therefore displaying great advantage to overcome the lingering nephrotoxicity of platinum anticancer drugs. Discrepant effects of a metal complex on mitochondria of different cells mean that targeting mitochondria has special significance in cancer therapy.
Collapse
Affiliation(s)
- Suxing Jin
- School
of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, P. R. China
- State
Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Yafeng He
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Chenyao Feng
- State
Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Jian Yuan
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Yan Guo
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Zijian Guo
- State
Key Laboratory of Coordination Chemistry, School of Chemistry and
Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Xiaoyong Wang
- State
Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
45
|
Calles A, Alonso M, Martín-Martorell P, Gómez A, de Castro J, Martínez-Aguillo M, Estival A, Mosquera J, Martínez-Banaclocha N, Majem M, Reyes R, Azkona E, Ortega AL, Aguin S, Santos A, Aguilar A, Cucurull M, Blasco A, Calvo V, Isla D, Nadal E, Aguado C, Sais E, Juan-Vidal O, Diz-Taín MP, Taus Á, Villanueva N, Bayona C, Amenedo M, Mielgo X, Arriola E, Baena J, Spanish Lung Cancer Group. Efficacy and safety of lorlatinib in patients with ALK- and ROS1-rearranged metastatic non-small cell lung cancer treated within the compassionate use program in Spain. Cancer Treat Res Commun 2025; 43:100905. [PMID: 40154161 DOI: 10.1016/j.ctarc.2025.100905] [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: 11/03/2024] [Revised: 01/21/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Lorlatinib, a third-generation tyrosine kinase inhibitor (TKI), targets both ALK and ROS1 rearrangements in non-small cell lung cancer (NSCLC). It is approved for ALK-positive patients after progression on prior TKIs but lacks FDA or EMA approval for ROS1-positive NSCLC. This study evaluates lorlatinib's efficacy and safety in both ALK- and ROS1-positive patients through a compassionate use program in Spain. METHODS We analyzed ALK-positive patients treated from November 2016 to February 2019 and ROS1-positive patients treated from November 2016 to March 2021. Eligible patients had Stage IV NSCLC with confirmed ALK or ROS1 rearrangements and prior TKI therapy. For ALK-positive patients, at least two prior TKIs were required if crizotinib was used first. For ROS1-positive patients, prior crizotinib was required. RESULTS In 61 ALK-positive patients, 59 % had brain metastasis, and 85.2 % received at least two prior ALK TKIs. The overall response rate (ORR) was 32.8 %, with a median progression-free survival (PFS) of 11.2 months. Intracranial ORR was 47.6 %, with higher efficacy in patients with evaluable brain metastasis. In patients with 1, 2, or ≥3 lines of previous TKIs, we observed a median PFS of 15.1, 11.1 and 7.6 months, respectively. Among 42 ROS1-positive patients, 59 % had brain metastasis, and 61.9 % received ≥2 prior therapies. The confirmed ORR was 47.6 %, with 16.7 % complete responses. Median PFS was 10 months. Patients receiving crizotinib alone had a median PFS of 10 months, while those with two prior TKIs had a median PFS of 8.5 months. Intracranial response was 44.4 %, rising to 57.1 % in patients evaluable with brain metastasis. No new safety signals were observed. CONCLUSION Lorlatinib demonstrated consistent efficacy and manageable safety in both ALK- and ROS1-positive NSCLC patients treated under the compassionate use program in Spain. These real-world findings support its use as an effective treatment option in heavily pretreated patients. MICROABSTRACT We evaluated the efficacy and safety of lorlatinib in ALK- and ROS1-positive NSCLC patients within a compassionate use program in Spain. Among 61 ALK-positive patients, including 59 % with brain metastasis and 85.2 % treated with at least 2 prior ALK TKIs, lorlatinib achieved a confirmed overall response rate (ORR) of 32.8 % and a median progression-free survival (PFS) of 11.2 months. In 42 ROS1-positive patients previously treated with crizotinib, lorlatinib showed an ORR of 47.6 % and a median PFS of 10 months, confirming its clinical activity despite the lack of FDA or EMA approval for this indication.
Collapse
Affiliation(s)
- Antonio Calles
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| | - Mirian Alonso
- Medical Oncology Department, Hospital Virgen del Rocío, Sevilla, Spain
| | | | - Ana Gómez
- Medical Oncology Department, H. Ramón y Cajal, Madrid, Spain
| | | | | | - Anna Estival
- Medical Oncology Department, H. U. Insular de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Joaquin Mosquera
- Medical Oncology Department, Hospital Teresa Herrera, A Coruña, Spain
| | | | - Margarita Majem
- Medical Oncology Department, H. Santa Creu i Sant Pau, Barcelona, Spain
| | - Roxana Reyes
- Medical Oncology Department, H. Clínic Barcelona, Barcelona, Spain
| | - Eider Azkona
- Medical Oncology Department, Hospital Universitario Cruces, Baracaldo, Spain
| | | | - Santiago Aguin
- Medical Oncology Department, H. Santiago, Santiago de Compostela, Spain
| | - Ana Santos
- Medical Oncology Department, Complejo H. de Toledo, Toledo, Spain
| | - Andrés Aguilar
- Medical Oncology Department, H. Quirón-Dexeus, Barcelona, Spain
| | - Marc Cucurull
- Medical Oncology Department, ICO Badalona, Badalona, Spain
| | - Ana Blasco
- Medical Oncology Department, H. General de Valencia, Valencia, Spain
| | - Virginia Calvo
- Medical Oncology Department, H. Puerta de Hierro, Majadahonda, Spain
| | - Dolores Isla
- Medical Oncology Department, H. Lozano Blesa, Zaragoza, Spain
| | - Ernest Nadal
- Medical Oncology Department, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL) L'Hospitalet, Barcelona, Spain
| | - Carlos Aguado
- Medical Oncology Department, Hospital Clínico San Carlos, Madrid, Spain
| | - Elia Sais
- Medical Oncology Department, ICO Girona, Girona, Spain
| | | | - MPilar Diz-Taín
- Medical Oncology Department, Complejo Asistencial Universitario de León, Spain
| | - Álvaro Taus
- Medical Oncology Department, Hospital del Mar, Barcelona, Spain
| | | | | | - Margarita Amenedo
- Medical Oncology Department, Centro Onc. Reg. Galicia, A Coruña, Spain
| | - Xabier Mielgo
- Medical Oncology Department, H. U. Alcorcón, Alcorcón, Madrid, Spain
| | | | - Javier Baena
- Medical Oncology Department, H. 12 de Octubre, Madrid, Spain
| | | |
Collapse
|
46
|
Martín F, Alcon C, Marín E, Morales-Sánchez P, Manzano-Muñoz A, Díaz S, García M, Samitier J, Lu A, Villanueva A, Reguart N, Teixido C, Montero J. Novel selective strategies targeting the BCL-2 family to enhance clinical efficacy in ALK-rearranged non-small cell lung cancer. Cell Death Dis 2025; 16:194. [PMID: 40113795 PMCID: PMC11926089 DOI: 10.1038/s41419-025-07513-3] [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: 02/21/2024] [Revised: 01/29/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
ALK (anaplastic lymphoma kinase) rearrangements represent the third most predominant driver oncogene in non-small cell lung cancer (NSCLC). Although ALK inhibitors are the tyrosine kinase inhibitors (TKIs) with the longest survival rates in lung cancer, the complex systemic clinical evaluation and the apoptotic cell death evasion of drug-tolerant persister (DTP) cancer cells may limit their therapeutic response. We found that dynamic BH3 profiling (DBP) presents an excellent predictive capacity to ALK-TKIs, that would facilitate their use in a clinical setting and complementing the readout of standard diagnostic assays. In addition, we revealed novel acute adaptive mechanisms in response to ALK inhibitors in cell lines and patient-derived tumor cells. Consistently, all our cell models confirmed a rapid downregulation of the sensitizer protein NOXA, leading to dependence on the anti-apoptotic protein MCL-1 after treatment with ALK-TKIs. In some cases, the anti-apoptotic protein BCL-xL may contribute equally to this anti-apoptotic response. Importantly, these acute dependencies could be prevented with BH3 mimetics in vitro and in vivo, blocking tumor adaptation to treatment. Finally, we also demonstrated how dual reactivation of PI3K/AKT and MAPK signaling pathways can impair lorlatinib response, which could be overcome with specific inhibitors of both signaling pathways. In conclusion, our findings propose several therapeutic combinations that should be explored in future clinical trials to enhance ALK inhibitors efficacy and improve the clinical response in a broad NSCLC patient population.
Collapse
Affiliation(s)
- Fernando Martín
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Clara Alcon
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Elba Marín
- Division of Medical Oncology, Hospital Clínic, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Unitat funcional de Tumors Toràcics, Hospital Clínic, Barcelona, Spain
| | - Paula Morales-Sánchez
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Albert Manzano-Muñoz
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Sherley Díaz
- Department of Pathology and CORE Molecular Biology Laboratory, Hospital Clínic, Barcelona, Spain
| | - Mireia García
- Department of Pathology and CORE Molecular Biology Laboratory, Hospital Clínic, Barcelona, Spain
| | - Josep Samitier
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
- Department of Electronics and Biomedical Engineering, Faculty of Physics, University of Barcelona, Barcelona, Spain
| | - Albert Lu
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Alberto Villanueva
- Chemoresistance and Predictive Factors Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - Noemí Reguart
- Division of Medical Oncology, Hospital Clínic, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Unitat funcional de Tumors Toràcics, Hospital Clínic, Barcelona, Spain
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Cristina Teixido
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Unitat funcional de Tumors Toràcics, Hospital Clínic, Barcelona, Spain
- Department of Pathology and CORE Molecular Biology Laboratory, Hospital Clínic, Barcelona, Spain
- Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Joan Montero
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
47
|
Iso H, Yomota M, Shirakura Y, Yoshinaga T, Kawai S, Narita K, Seike M, Hosomi Y. Clinical Impact of Osimertinib Dose Reduction in the First-Line Setting on EGFR Mutation-Positive Non-Small Cell Lung Cancer: A Retrospective Monocentric Study. Onco Targets Ther 2025; 18:379-387. [PMID: 40124926 PMCID: PMC11930247 DOI: 10.2147/ott.s494112] [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: 09/13/2024] [Accepted: 03/04/2025] [Indexed: 03/25/2025] Open
Abstract
Purpose Osimertinib is a third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor. In the FLAURA trial, osimertinib demonstrated longer progression-free survival (PFS) and overall survival (OS) compared to gefitinib or erlotinib. In the trial, dose reductions occurred in 5% of patients, primarily due to QT prolongation. However, various adverse events can also lead to dose reductions in clinical practice, and the efficacy of osimertinib after dose reduction remains unclear. The present study was conducted to evaluate the clinical impact of osimertinib dose reduction. Patients and Methods This monocentric retrospective study was conducted at Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital. Ninety patients with EGFR mutation-positive non-squamous non-small-cell lung cancer receiving osimertinib as their first-line therapy between August 2018 and December 2021 were included. Results Of the cohort, 23 patients had an osimertinib dose reduction during their clinical course. The dose reduction group tended to have a lower median body weight and a higher proportion of elderly patients aged 80 years or older. The median PFS was 21.2 months (95% confidence interval [CI]: 8.22-34.18) in the dose reduction group and 18.6 (95% CI: 13.04-24.23) months in the regular-dose group. The median OS was 29.6 months (95% CI: 17.44-41.70) in the osimertinib dose-reduction group and 37.7 (95% CI: 27.10-48.23) months in the regular-dose group. Dose reduction did not significantly impact the time-dependent hazard ratio (HR) for PFS (HR 1.22 [95% CI: 0.55-1.89]) or OS (HR: 1.24 [95% CI: 0.64-2.42]). The adverse events leading to dose reduction were mainly rash, anorexia, and paronychia, and no fatal adverse events were observed after dose reduction. Conclusion The present study suggests that dose reduction may not compromise the efficacy of osimertinib. However, the clinical impact of dose reduction is not fully understood. Physicians should carefully weigh its benefits and risks before implementation.
Collapse
Affiliation(s)
- Hirokazu Iso
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Makiko Yomota
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Yukari Shirakura
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Tadatsugu Yoshinaga
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Shoko Kawai
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Kosuke Narita
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Masahiro Seike
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Yukio Hosomi
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| |
Collapse
|
48
|
Sattler M, Salgia R. The expanding role of the receptor tyrosine kinase MET as a therapeutic target in non-small cell lung cancer. Cell Rep Med 2025; 6:101983. [PMID: 40020676 PMCID: PMC11970332 DOI: 10.1016/j.xcrm.2025.101983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 03/03/2025]
Abstract
Aberrant regulation of MET receptor tyrosine kinase activity is a frequent event in non-small cell lung cancer (NSCLC), even though the frequency of oncogenic driver mutations of MET is low. Our discovery of oncogenic MET exon 14 skipping mutations, the characterization of the first prototype MET kinase inhibitor, and characterization of MET expression levels have led the way to novel therapeutic approaches with improved outcomes in NSCLC. MET exon 14 mutations are the most consequential but not the only alterations that can be targeted through small molecule tyrosine kinase inhibitors. The abundant expression of cellular MET (c-MET) in cancer cells has provided new opportunities for immuno-oncology approaches in a broader patient population, and the integration of MET-targeted personalized medicine with immunotherapy has not been fully exploited yet. Here, we highlight essential facets of MET as a therapeutic target in NSCLC and provide an outlook for future approaches.
Collapse
Affiliation(s)
- Martin Sattler
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
49
|
Zhang Y, Xu Y, Jin H, Liu T, Zhong H, Xu J, Lou Y, Zhong R. Spatial Heterogeneity of PD-L1 Expression as a Biomarker for Third-Generation EGFR-TKI Response in Advanced EGFR-Mutant NSCLC. Cancer Sci 2025. [PMID: 40102299 DOI: 10.1111/cas.70060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/07/2025] [Accepted: 03/13/2025] [Indexed: 03/20/2025] Open
Abstract
The association between the spatial heterogeneity of programmed cell death ligand 1 (PD-L1) expression and the efficacy of third-generation epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) in EGFR-mutant non-small cell lung cancer (NSCLC) remains elusive. This retrospective study analyzed data from 4171 NSCLC patients with EGFR-sensitive mutations treated at Shanghai Chest Hospital from August 2019 to September 2023. Among them, 182 patients receiving third-generation EGFR-TKIs monotherapy as a first-line treatment were enrolled. Patients were categorized by biopsy sites into primary lung lesions (n = 112) and metastatic lymph nodes (n = 70). PD-L1 expression was stratified based on tumor cell proportion score (TPS): < 1%, 1%-49%, and ≥ 50%. The median progression-free survival (PFS) for the entire cohort was 18.33 months. In the PD-L1 TPS group, PFS was 18.87 months for TPS < 1%, 17.6 months for TPS 1%-49%, and 13.6 months for TPS ≥ 50%, with significant differences across groups (p = 0.026). Moreover, multivariate analysis identified smoking history [HR = 1.653, 95% CI (1.132-2.414), p = 0.009] and TPS ≥ 50% [HR = 2.069, 95% CI (1.183-3.618), p = 0.011] as independent risk factors. In primary lesions, the median PFS was 21.93 months for TPS < 1%, 18.57 months for TPS 1%-49%, and 10.17 months for TPS ≥ 50%, with significant differences (p < 0.001). However, PD-L1 expression in metastatic lymph nodes was not associated with PFS (p = 0.973). In advanced EGFR-mutant NSCLC, high PD-L1 expression may suggest reduced efficacy of third-generation EGFR-TKIs. The spatial heterogeneity of PD-L1 expression could influence its predictive accuracy for third-generation EGFR-TKI efficacy.
Collapse
Affiliation(s)
- Yidan Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yingqi Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongping Jin
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tengfei Liu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Zhong
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianlin Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuqing Lou
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Runbo Zhong
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
50
|
Parisi C, Planchard D. BRAF in non-small cell lung cancer: From molecular mechanisms to clinical practice. Cancer 2025; 131 Suppl 1:e35781. [PMID: 40172088 DOI: 10.1002/cncr.35781] [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: 08/26/2024] [Revised: 10/30/2024] [Accepted: 11/21/2024] [Indexed: 04/04/2025]
Abstract
V-Raf murine sarcoma viral oncogene homolog B (BRAF) mutations are found in up to 4% of patients with non-small cell lung cancer (NSCLC). Approximately 2% of advanced NSCLC cases harbor a BRAF V600E (class I) mutation. Because targeted therapies inhibiting BRAF (e.g., dabrafenib and encorafenib) and MEK (trametinib and binimetinib) are associated with improved outcomes as first- or second-line treatment for BRAF V600E-mutant NSCLC, both European Society for Medical Oncology and National Comprehensive Cancer Network guidelines recommend testing for the BRAF V600E oncogenic driver at the time of diagnosis. In recent years, the treatment landscape of this molecular subgroup has seen great development. Different therapeutic strategies including anti-programmed death ligand 1 antibodies and kinase inhibitors have been assessed thus far, with novel agents (e.g., pan-BRAF inhibitors) and therapeutic associations underway in preclinical and clinical trials. This review describes the current understanding of the BRAF clinicopathologic role in NSCLC, with a special focus on published trials assessing currently approved therapies. Mechanisms of drug resistance and future perspectives on the therapeutic approach of BRAF-deregulated NSCLC are also summarized.
Collapse
Affiliation(s)
- Claudia Parisi
- Thoracic Cancer Group, Department of Medical Oncology, Gustave Roussy and International Center for Thoracic Cancers, Villejuif, France
- Paris-Saclay University, Villejuif, Kremlin-Bicêtre, France
- Department of Medical-Surgical Sciences and Translational Medicine, Sapienza University of Rome, Rome, Italy
| | - David Planchard
- Thoracic Cancer Group, Department of Medical Oncology, Gustave Roussy and International Center for Thoracic Cancers, Villejuif, France
- Paris-Saclay University, Villejuif, Kremlin-Bicêtre, France
| |
Collapse
|