1
|
Gaeta B, Eichholz JE, Walch H, Ilica AT, Boe L, Kratochvil L, Yu Y, Gomez DR, Imber BS, Li BT, Murciano-Goroff YR, Arbour KC, Schultz N, Lebow ES, Pike LRG. Intracranial Disease Control and Survival among Patients with KRAS-mutant Lung Adenocarcinoma and Brain Metastases Treated with Stereotactic Radiosurgery. Int J Radiat Oncol Biol Phys 2025; 122:424-434. [PMID: 39929348 DOI: 10.1016/j.ijrobp.2025.01.033] [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: 08/06/2024] [Revised: 01/14/2025] [Accepted: 01/25/2025] [Indexed: 03/01/2025]
Abstract
PURPOSE Precision medicine according to molecularly defined subgroups offers great potential to improve outcomes for patients with metastatic lung adenocarcinoma. This study describes clinical outcomes and the impact of co-occurring genetic alterations on outcomes following stereotactic radiosurgery (SRS) among patients with Kirsten rat sarcoma viral oncogene homolog (KRAS)-mutant lung adenocarcinoma. METHODS AND MATERIALS A total of 195 patients with KRAS-mutant lung adenocarcinoma were treated with SRS for brain metastases (BMs) between 2014 and 2018 with follow-up until 2022 or death. Coprimary outcomes were median overall survival (OS) and intracranial progression-free survival (iPFS); univariable and multivariable Cox regression models and Kaplan-Meier survival analysis were used. RESULTS Median follow-up from the date of BM diagnosis was 11 months. Median OS and iPFS for the cohort were 27.7 months (95% CI, 19.7-36.8) and 22.1 months (95% CI, 16.8-48.9), respectively. Lesion-level local control at 12 and 24 months was 89.9% and 87.5%, respectively. In a multivariable Cox regression model, inferior OS was associated with coalterations in KEAP1 and STK11 (hazard ratio [HR], 1.94; 95% CI, 1.04-3.62; q = 0.087), progressive (HR, 3.41; 95% CI, 1.38-8.39; q = 0.087), and mixed response (HR, 3.52; 95% CI, 1.2-10.3; q = 0.092) extracranial disease, and 6 or more BMs at time of diagnosis (HR, 2.58; 95% CI, 1.22-6.63; q = 0.087). Positive programmed death ligand 1 status was associated with improved OS (HR, 0.57; 95% CI, 0.37-0.87; P = .01). Inferior iPFS was associated with chemotherapy before SRS (HR, 2.69; 95% CI, 1.42-5.09; q = 0.04) and age >65 years (HR, 2.21; 95% CI, 1.25-3.93; q = 0.055). KRAS G12C was not associated with differences in iPFS, OS, or type of intracranial progression event following SRS. CONCLUSIONS Coalteration of KRAS and KEAP1/STK11 was associated with inferior OS, but not iPFS. Similar outcomes were found in patients harboring KRAS G12C and non-G12C mutant non-small cell lung cancer BMs. Further understanding of molecularly characterized subgroups will be critical in driving personalized radiation therapy for patients with lung cancer BMs.
Collapse
Affiliation(s)
- Benjamin Gaeta
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Jordan E Eichholz
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Henry Walch
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmet T Ilica
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lillian Boe
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Leah Kratochvil
- Memorial Hospital Research Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yao Yu
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniel R Gomez
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Global Biomarker Development Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Brandon S Imber
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Bob T Li
- Memorial Hospital Research Program, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yonina R Murciano-Goroff
- Memorial Hospital Research Program, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kathryn C Arbour
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nikolaus Schultz
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Emily S Lebow
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Global Biomarker Development Program, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Radiation Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Luke R G Pike
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York; Global Biomarker Development Program, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
2
|
Rao W, Zhang Q, Dai X, Yang Y, Lei Z, Kuang X, Xiao H, Zhu J, Xiong Y, Wang D, Yang L. A three-subtype prognostic classification based on base excision repair and oxidative stress genes in lung adenocarcinoma and its relationship with tumor microenvironment. Sci Rep 2025; 15:16647. [PMID: 40360689 PMCID: PMC12075871 DOI: 10.1038/s41598-025-98088-8] [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: 10/23/2024] [Accepted: 04/09/2025] [Indexed: 05/15/2025] Open
Abstract
Unrepaired DNA damage is the initiation of mutation and tumor-specific biological characteristics. Oxidative stress and base excision repair (BER) are the two main pathways to cope with oxidative DNA damage, which is closely related to the heterogeneity of Lung adenocarcinoma (LUAD), but their relationship with tumor biological characteristics is unclear, and a molecular subtyping based on comprehensive BER and oxidative stress gene expression is lacking. 501 samples from The Cancer Genome Atlas (TCGA) were classified into three subtypes based on genes related to BER and oxidative stress through hierarchical agglomerative cluster analysis. By integrating the nearest template prediction (NTP), four GEO datasets and 52 samples from our institution were analyzed for validation. Bioinformatic analysis was performed to define the diverse molecular characteristics, mutation background, tumor microenvironment, and prognosis. Three subtypes with distinct gene signatures were identified: relatively high BER and low oxidative stress gene expression (C1), low BER gene and high oxidative stress gene expression (C2), and high expression of both BER and oxidative stress genes (C3). C2 was characterized by a low mutation frequency in TP53 (29%) and a high mutation frequency in EGFR (20%), whereas a high frequency of mutation was seen in C3 in STK11 and KEAP1 genes. Additionally, differentially expressed genes among the three subtypes were particularly enriched in immune-related pathways, and the abundance of immune cells and Immunophenoscore were significantly higher in C2, while the Tumor Immune Dysfunction and Exclusion (TIDE) score was lower in C2, indicating a better response to immunotherapy. C2 was also associated with an improved survival outcome compared with C1 and C3, and this finding was validated in 978 samples from four independent GEO datasets and 52 samples at our institution by the NTP algorithm. The three-subtype classifications based on BER and oxidative stress gene expression offers potential for predicting the survival and response to immunotherapy of LUAD patients.
Collapse
Affiliation(s)
- Wen Rao
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
- The 75th Group Army Hospital, Dali, Yunnan, People's Republic of China
| | - Qin Zhang
- Department of Radiation Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Cancer Hospital Affiliate to School of Medicine,, University of Electronic Science and Technology of China, Sichuan, People's Republic of China
| | - Xiaoyan Dai
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
| | - Yuxin Yang
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
| | - Zhang Lei
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
| | - Xunjie Kuang
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
| | - He Xiao
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
| | - Jianwu Zhu
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
| | - Yanli Xiong
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China
| | - Dong Wang
- Chongqing University Qianjiang Hospital, Chongqing, People's Republic of China.
| | - Lujie Yang
- Cancer Center, Daping Hospital and Army Medical Center of PLA, Army Medical University, No.10 Changjiangzhi Rd, Yuzhong District, Chongqing, People's Republic of China.
| |
Collapse
|
3
|
Gomatou G, Charpidou A, Li P, Syrigos N, Gkiozos I. Mechanisms of primary resistance to immune checkpoint inhibitors in NSCLC. Clin Transl Oncol 2025; 27:1426-1437. [PMID: 39307892 DOI: 10.1007/s12094-024-03731-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/10/2024] [Indexed: 04/16/2025]
Abstract
Immune checkpoint inhibitors (ICIs) redefined the therapeutics of non-small cell lung cancer (NSCLC), leading to significant survival benefits and unprecedented durable responses. However, the majority of the patients develop resistance to ICIs, either primary or acquired. Establishing a definition of primary resistance to ICIs in different clinical scenarios is challenging and remains a work in progress due to the changing landscape of ICI-based regimens, mainly in the setting of early-stage NSCLC. The mechanisms of primary resistance to ICIs in patients with NSCLC include a plethora of pathways involving a cross-talk of the tumor cells, the tumor microenvironment and the host, leading to the development of an immunosuppressive phenotype. The optimal management of patients with NSCLC following primary resistance to ICIs represents a significant challenge in current thoracic oncology. Research in this field includes exploring other immunotherapeutic approaches, such as cancer vaccines, and investigating novel antibody-drug conjugates in patients with NSCLC.
Collapse
Affiliation(s)
- Georgia Gomatou
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Chest Diseases, National and Kapodistrian University of Athens, Athens, Greece.
| | - Andriani Charpidou
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Chest Diseases, National and Kapodistrian University of Athens, Athens, Greece
| | - Peifeng Li
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Nikolaos Syrigos
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Chest Diseases, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Gkiozos
- Oncology Unit, Third Department of Medicine, "Sotiria" General Hospital for Chest Diseases, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
4
|
Cuadrado A, Cazalla E, Bach A, Bathish B, Naidu SD, DeNicola GM, Dinkova-Kostova AT, Fernández-Ginés R, Grochot-Przeczek A, Hayes JD, Kensler TW, León R, Liby KT, López MG, Manda G, Shivakumar AK, Hakomäki H, Moerland JA, Motohashi H, Rojo AI, Sykiotis GP, Taguchi K, Valverde ÁM, Yamamoto M, Levonen AL. Health position paper and redox perspectives - Bench to bedside transition for pharmacological regulation of NRF2 in noncommunicable diseases. Redox Biol 2025; 81:103569. [PMID: 40059038 PMCID: PMC11970334 DOI: 10.1016/j.redox.2025.103569] [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: 12/21/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a redox-activated transcription factor regulating cellular defense against oxidative stress, thereby playing a pivotal role in maintaining cellular homeostasis. Its dysregulation is implicated in the progression of a wide array of human diseases, making NRF2 a compelling target for therapeutic interventions. However, challenges persist in drug discovery and safe targeting of NRF2, as unresolved questions remain especially regarding its context-specific role in diseases and off-target effects. This comprehensive review discusses the dualistic role of NRF2 in disease pathophysiology, covering its protective and/or destructive roles in autoimmune, respiratory, cardiovascular, and metabolic diseases, as well as diseases of the digestive system and cancer. Additionally, we also review the development of drugs that either activate or inhibit NRF2, discuss main barriers in translating NRF2-based therapies from bench to bedside, and consider the ways to monitor NRF2 activation in vivo.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Eduardo Cazalla
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anders Bach
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Boushra Bathish
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Sharadha Dayalan Naidu
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Gina M DeNicola
- Department of Metabolism and Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Raquel Fernández-Ginés
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Thomas W Kensler
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28007, Madrid, Spain
| | - Karen T Liby
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Manuela G López
- Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain; Instituto Teófilo Hernando, Madrid, Spain
| | - Gina Manda
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | | | - Henriikka Hakomäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jessica A Moerland
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Hozumi Motohashi
- Department of Medical Biochemistry, Graduate School of Medicine Tohoku University, Sendai, Japan; Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ana I Rojo
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Keiko Taguchi
- Laboratory of Food Chemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan; Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas "Sols-Morreale" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
5
|
Di Federico A, Hong L, Elkrief A, Thummalapalli R, Cooper AJ, Ricciuti B, Digumarthy S, Alessi JV, Gogia P, Pecci F, Makarem M, Gandhi MM, Garbo E, Saini A, De Giglio A, Favorito V, Scalera S, Cipriani L, Marinelli D, Haradon D, Nguyen T, Haradon J, Voligny E, Vaz V, Gelsomino F, Sperandi F, Melotti B, Ladanyi M, Zhang J, Gibbons DL, Heymach JV, Nishino M, Lindsay J, Rodig SJ, Pfaff K, Sholl LM, Wang X, Johnson BE, Jänne PA, Rekhtman N, Maugeri-Saccà M, Heist RS, Ardizzoni A, Awad MM, Arbour KC, Schoenfeld AJ, Vokes NI, Luo J. Lung adenocarcinomas with mucinous histology: clinical, genomic, and immune microenvironment characterization and outcomes to immunotherapy-based treatments and KRAS G12C inhibitors. Ann Oncol 2025; 36:297-308. [PMID: 39637943 PMCID: PMC11845285 DOI: 10.1016/j.annonc.2024.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Approximately 10% of lung adenocarcinomas (LUADs) have mucinous histology (LUADMuc), which is associated with a light/absent smoking history and a high prevalence of KRAS mutations. We sought to characterize LUADMuc by comparing it with LUAD without mucinous histology (LUADnon-muc) and determine the relative benefit of current treatments. PATIENTS AND METHODS Patients with LUAD from five institutions and The Cancer Genome Atlas Pan-Cancer Atlas classified as LUADMuc or LUADnon-muc were included. Clinicopathologic, genomic, immunophenotypic, transcriptional features, and treatment outcomes were compared between LUADMuc and LUADnon-muc. RESULTS Of 4082 patients with LUAD, 9.9% had LUADMuc. Compared with LUADnon-muc, patients with LUADMuc had a lighter smoking history (median 15 versus 20 pack-years; P = 0.008), lower programmed death-ligand 1 (PD-L1) tumor proportion score (median 0% versus 5%, P < 0.0001), and lower tumor mutation burden (median 6.8 versus 8.5 mutations/megabase, P < 0.0001). Mutations in KRAS, NKX2-1 [thyroid transcription factor 1 (TTF-1)], STK11, SMARCA4, GNAS, and ALK rearrangements were enriched in LUADMuc, while TP53, EGFR, BRAF, and MET mutations were enriched in LUADnon-muc. At stage IV diagnosis, LUADMuc was more likely to have contralateral lung metastasis (55.2% versus 36.9%, P < 0.0001) and less likely to have brain metastases (23.3% versus 41.9%, P < 0.0001). Compared with LUADnon-muc, LUADMuc cases showed lower intratumor CD8+, PD-1+, CD8+PD-1+, and FOXP3+ cells. Among metastatic cases receiving immune checkpoint inhibitors, compared with LUADnon-muc (n = 1511), LUADMuc (n = 112) had a lower objective response rate (ORR 8.4% versus 25.9%, P < 0.0001), and shorter median progression-free survival (mPFS 2.6 versus 3.9 months, P < 0.0001) and overall survival (mOS 9.9 versus 17.2 months, P < 0.0001). Similarly, patients with LUADMuc had worse outcomes to chemoimmunotherapy. LUADMuc (n = 18) and LUADnon-muc (n = 150) had similar ORR (16.7% versus 34.9%, P = 0.12) and mPFS (4.6 versus 5.6 months, P = 0.17) to treatment with KRASG12C inhibitors, but LUADMuc had shorter mOS (6.8 versus 10.8 months, P = 0.018). CONCLUSIONS LUADMuc represents a distinct LUAD subpopulation with unique clinicopathologic, genomic, immunophenotypic, and transcriptional features, achieving worse outcomes to standard immunotherapy-based treatments.
Collapse
Affiliation(s)
- A Di Federico
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston
| | - L Hong
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - A Elkrief
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - R Thummalapalli
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - A J Cooper
- Massachusetts General Hospital, Boston, USA
| | - B Ricciuti
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston
| | | | - J V Alessi
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston
| | - P Gogia
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - F Pecci
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston
| | - M Makarem
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston
| | - M M Gandhi
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston
| | - E Garbo
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston
| | - A Saini
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston
| | - A De Giglio
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna
| | - V Favorito
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna
| | - S Scalera
- Clinical Trial Center, Biostatistics and Bioinformatics Division, IRCCS Regina Elena National Cancer Institute, Rome
| | - L Cipriani
- Clinical Trial Center, Biostatistics and Bioinformatics Division, IRCCS Regina Elena National Cancer Institute, Rome
| | - D Marinelli
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - D Haradon
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston
| | - T Nguyen
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston
| | - J Haradon
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston
| | - E Voligny
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston
| | - V Vaz
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston
| | - F Gelsomino
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna
| | - F Sperandi
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna
| | - B Melotti
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna
| | - M Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - J Zhang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - D L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - J V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - M Nishino
- Department of Radiology, Brigham and Women's Hospital, Boston; Department of Imaging, Dana-Farber Cancer Institute, Boston
| | - J Lindsay
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston
| | - S J Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston
| | - K Pfaff
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston
| | - L M Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston
| | - X Wang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, Boston
| | - B E Johnson
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| | - P A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| | - N Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York
| | - M Maugeri-Saccà
- Clinical Trial Center, Biostatistics and Bioinformatics Division, IRCCS Regina Elena National Cancer Institute, Rome
| | - R S Heist
- Massachusetts General Hospital, Boston, USA
| | - A Ardizzoni
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna
| | - M M Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, USA
| | - K C Arbour
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - A J Schoenfeld
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York
| | - N I Vokes
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - J Luo
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, USA.
| |
Collapse
|
6
|
Xu L, Xu X, Wu P, Ye W, Zhao J, Yang J, Yao Y, Chen M, Wang X, Wang A, Fan Y. Clinical characteristics and prognostic analysis of patients with SMARCA4-deficient lung cancer. Technol Health Care 2025; 33:1014-1020. [PMID: 40105165 DOI: 10.1177/09287329241296242] [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: 03/20/2025]
Abstract
BackgroundSMARCA4-deficient NSCLC is a rare type of tumor, accounting for approximately 10% of all NSCLC. It exhibits a weak response to conventional chemotherapy and has a poor prognosis, and lacks alterations in EGFR (epidermal growth factor receptor), ALK (anaplastic lymphoma kinase), and ROS1 (ROS proto-oncogene 1) genes Therefore, the mechanisms of SMARCA4 in NSCLC development urgently need to be explored to identify novel biomarkers and precise therapeutic strategies for this subtype.ObjectiveThe aim of this study was to understand the clinical characteristics of this special type of tumor and its response to different treatments.MethodsWe collected clinical data from 42 patients with SMARCA4-deficient NSCLC from July 2022 to January 2024, and analyzed their clinical features and survival state.ResultsThe study included a total of 42 patients diagnosed with NSCLC and harboring SMARCA4 mutation. The majority of these patients were male with a median age of 67 years. Most patients presented at stage IV upon diagnosis with highly aggressive tumors characterized by high Ki-67 proliferation index values resulting in poor overall prognosis. Genetic testing revealed TP53 gene mutations to be most prevalent (21%), followed by KRAS mutations (13%). Patients receiving immunotherapy exhibited significantly longer median overall survival compared to those treated solely with chemotherapy. Targeted drug therapy demonstrated favorable effects in some patients.ConclusionNSCLC patients harboring SMARCA4 deficiency exhibit poor overall survival rates with a median overall survival time of 5.4 months. Immunotherapy may provide benefits for NSCLC patients with SMARCA4 deficiency.
Collapse
Affiliation(s)
- Lingling Xu
- Department of Oncology, Anhui Chest Hospital, Hefei, China
| | - Xianquan Xu
- Thoracic Surgery, Anhui Chest Hospital, Hefei, China
| | - Pengfei Wu
- Department of Oncology, Anhui Chest Hospital, Hefei, China
| | - Wei Ye
- Pathology Department, Anhui Chest Hospital, Hefei, China
| | - Jieting Zhao
- Pathology Department, Anhui Chest Hospital, Hefei, China
| | - Jingwen Yang
- Department of Oncology, Anhui Chest Hospital, Hefei, China
| | - Yuanyuan Yao
- Department of Oncology, Anhui Chest Hospital, Hefei, China
| | - Maoxi Chen
- Department of Oncology, Anhui Chest Hospital, Hefei, China
| | - Xiaoyan Wang
- Department of Oncology, Anhui Chest Hospital, Hefei, China
| | - Anbang Wang
- Department of Oncology, Anhui Chest Hospital, Hefei, China
| | - Yanbo Fan
- Department of Oncology, Anhui Chest Hospital, Hefei, China
| |
Collapse
|
7
|
Ge Y, Zhou Q, Pan F, Wang R. Utilizing Nanoparticles to Overcome Anti-PD-1/PD-L1 Immunotherapy Resistance in Non-Small Cell Lung cancer: A Potential Strategy. Int J Nanomedicine 2025; 20:2371-2394. [PMID: 40027868 PMCID: PMC11871910 DOI: 10.2147/ijn.s505539] [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: 11/21/2024] [Accepted: 01/25/2025] [Indexed: 03/05/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality globally, with non-small cell lung cancer (NSCLC) constituting 85% of cases. Immune checkpoint inhibitors (ICIs) represented by anti-programmed cell death protein 1 (PD-1)/ programmed cell death ligand 1 (PD-L1) have emerged as a promising frontier in cancer treatment, effectively extending the survival of patients with NSCLC. However, the efficacy of ICIs exhibits significant variability across diverse patient populations, with a substantial proportion showing poor responsiveness and acquired resistance in those initially responsive to ICIs treatments. With the advancement of nanotechnology, nanoparticles offer unique advantages in tumor immunotherapy, including high permeability and prolonged retention(EPR) effects, enhanced drug delivery and stability, and modulation of the inflammatory tumor microenvironment(TME). This review summarizes the mechanisms of resistance to ICIs in NSCLC, focusing on tumor antigens loss and defective antigen processing and presentation, failure T cell priming, impaired T cell migration and infiltration, immunosuppressive TME, and genetic mutations. Furthermore, we discuss how nanoparticles, through their intrinsic properties such as the EPR effect, active targeting effect, shielding effect, self-regulatory effect, and synergistic effect, can potentiate the efficacy of ICIs and reverse resistance. In conclusion, nanoparticles serve as a robust platform for ICIs-based NSCLC therapy, aiding in overcoming resistance challenges.
Collapse
Affiliation(s)
- Yuli Ge
- Department of Medical Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| | - Qiong Zhou
- Department of Medical Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210093, People’s Republic of China
| | - Fan Pan
- Department of Medical Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, 210093, People’s Republic of China
| | - Rui Wang
- Department of Medical Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210023, People’s Republic of China
| |
Collapse
|
8
|
Yin D, Lu X, Liang X, Lu Y, Xiong L, Wu P, Wang T, Chen J. STK11 genetic alterations in metastatic EGFR mutant lung cancer. Sci Rep 2025; 15:5729. [PMID: 39962098 PMCID: PMC11832735 DOI: 10.1038/s41598-024-74779-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/30/2024] [Indexed: 02/20/2025] Open
Abstract
This study was conducted to investigate the relationship between STK11 genetic alterations and the outcomes of patients with metastatic EGFR mutant lung cancer. Clinical characteristics and genomic data were downloaded from the cBioPortal database. The information of the case with STK11 mutation was collected from Jiangyin People's Hospital. Univariate and multivariate analyses were performed to distinguish the prognostic differences. Outcomes were analyzed before and after propensity score matching (PSM). A patient with STK11 mutation was insensitive to osimertinib and had an extremely poor prognosis. Further analysis showed that STK11 mutations had a strong mutual exclusion with EGFR mutations. A total of 960 patients with metastatic EGFR mutant lung adenocarcinoma were enrolled in the prognostic analysis. STK11 alternation was a significant predictor of worse outcomes in univariate or multivariate analyses. After PSM, patients with STK11 alternations still exhibited poor prognoses. Cell culture experiments also showed that the loss of STK11 could contribute to the resistance of osimertinib. Functionally, STK11 mutation was positively associated with metabolic signaling pathways and immune infiltrates negatively. Through drug screening, trametinib was identified to sensitize osimertinib in the STK11-deficient cell. This study found that STK11 genetic alterations portend a worse prognosis for patients with metastatic EGFR mutant lung cancer and led to osimertinib resistance potentially. MEK inhibitors could sensitize osimertinib in the STK11-deficient cell.
Collapse
Affiliation(s)
- Dandan Yin
- The Second Hospital of Nanjing, Clinical Teaching Hospital of Medical School, Nanjing University, Nanjing, People's Republic of China
- Clinical Research Center, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210003, Jiangsu, People's Republic of China
| | - Xiyi Lu
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Xiao Liang
- Department of Oncology, The Affiliated Jiangyin Hospital of Nantong University, Wuxi, Jiangsu, People's Republic of China
| | - Yiting Lu
- Department of Radiology, The Affiliated Jiangyin Hospital of Nantong University, Wuxi, Jiangsu, People's Republic of China
| | - Lei Xiong
- Department of Cardiothoracic Surgery, Jinling Hospital, Jinling Clinical Medical School, Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China.
| | - Pingping Wu
- Department of Oncology, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institue of Cancer Research, Nanjing, Jiangsu, People's Republic of China.
| | - Tingting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, People's Republic of China.
- Medical School, Nanjing University, Nanjing, People's Republic of China.
| | - Jinfei Chen
- Medical School, Nanjing University, Nanjing, People's Republic of China.
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
9
|
Ramos-Ramírez M, Caballe-Pérez E, Lucio-Lozada J, Romero-Nuñez E, Castillo-Ruiz C, Dorantes-Sánchez L, Flores-Estrada D, Recondo G, Barrios-Bernal P, Cabrera-Miranda L, Bravo-Dominguez H, Hernández-Pedro N, Arrieta O. Immunomodulatory role of oncogenic alterations in non-small cell lung cancer: a review of implications for immunotherapy. Cancer Metastasis Rev 2025; 44:30. [PMID: 39915358 DOI: 10.1007/s10555-025-10245-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 01/16/2025] [Indexed: 03/28/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have improved clinical outcomes in patients with non-small cell lung cancer (NSCLC) lacking targetable oncogenic alterations. However, their efficacy in individuals with such genomic alterations remains heterogeneous and poorly understood. In detail, certain oncogenic alterations in TP53, EGFR (uncommon mutations), KRAS (G12C), BRAF (non-V600E), MET (amplifications), FGFR1 and FGFR4, actively modify MAPK, PI3K, and STING signaling, thus remodeling tumoral immune phenotype and are associated with high TMB counts, enriched T lymphocyte tumor infiltration, and high expression of antigen-presenting molecules, supporting their consideration as part of the eligibility criteria for ICIs treatment. Nonetheless, other oncogenic alterations are associated with an immunosuppressive TME, low TMB counts, and downregulation of targetable immune checkpoints, in which novel therapeutic approaches are currently being tested to overcome their intrinsic resistance. In this context, this review discusses the fundamental mechanisms by which frequent driver alterations affect ICIs efficacy in patients with NSCLC, and outlines their prognostic relevance in the era of immunotherapy.
Collapse
Affiliation(s)
- Maritza Ramos-Ramírez
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - Enrique Caballe-Pérez
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - José Lucio-Lozada
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Eunice Romero-Nuñez
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Cesar Castillo-Ruiz
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Lorena Dorantes-Sánchez
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Diana Flores-Estrada
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - Gonzalo Recondo
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Pedro Barrios-Bernal
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico
| | - Luis Cabrera-Miranda
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - Heyman Bravo-Dominguez
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico
| | - Norma Hernández-Pedro
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico.
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico.
| | - Oscar Arrieta
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN), Mexico City, (CDMX), Mexico.
- Thoracic Oncology Functional Unit (UFOT), Instituto Nacional de Cancerología, (INCAN), Mexico City , (CDMX), Mexico.
| |
Collapse
|
10
|
Numata T, Nakamura R, Shiozawa T, Watanabe H, Okauchi S, Ogara G, Tamura T, Kikuchi N, Miyazaki K, Hayashi S, Yamashita T, Kurishima K, Inagaki M, Satoh H, Kaburagi T, Endo T, Hizawa N. Outcomes of Combined Atezolizumab Plus Chemotherapy in Non-small Cell Lung Cancer Patients in Clinical Practice. CANCER DIAGNOSIS & PROGNOSIS 2025; 5:105-114. [PMID: 39758245 PMCID: PMC11696331 DOI: 10.21873/cdp.10418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 01/07/2025]
Abstract
Background/Aim Atezolizumab, one of the anti-PD-L1 antibodies, targets PD-L1 expressed on cancer cells and antigen-presenting cells. This immune checkpoint inhibitor is now commonly used in combination with chemotherapy. The objectives of this study were to confirm the treatment outcomes of combined atezolizumab plus chemotherapy, and to identify prognostic factors, with a particular focus on the impact of the site of metastasis in real-world clinical practice. Patients and Methods A retrospective review of clinical information on non-small cell lung cancer patients who received combined atezolizumab plus chemotherapy from May 2018 to August 2024 at our 11 hospitals was conducted. Results The 141 patients evaluated had a median progression-free survival of 8.0 months and a median overall survival of 19.0 months. Multivariate analyses showed that 'absence of liver metastases', 'absence of adrenal metastases', 'first-line combined atezolizumab plus chemotherapy', and 'good performance status' were associated with progression-free survival and overall survival. Immune-related adverse events were observed in 27.7% of patients, with grade 3 or higher in 9.9% of patients, and grade 5 in 2.1% of patients. Conclusion Efficacy and immune-related adverse events associated with the combination of atezolizumab and chemotherapy in non-small cell lung cancer patients were comparable to previous clinical trials. To ensure that appropriate patients receive the most effective treatment, it is important to identify detailed prognostic factors, including clinical information, such as the affected metastatic organs. Continued research and further accumulation of knowledge in this area are eagerly anticipated.
Collapse
Affiliation(s)
- Takeshi Numata
- Departments of Respiratory Medicine and Surgery, National Hospital Organization Mito Medical Center, Ibarakimachi, Japan
| | - Ryota Nakamura
- Departments of Respiratory Medicine and Surgery, National Hospital Organization Mito Medical Center, Ibarakimachi, Japan
| | - Toshihiro Shiozawa
- Division of Respiratory Medicine, Faculty of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hiroko Watanabe
- Division of Respiratory Medicine, Tsukuba Memorial Hospital, Tsukuba, Japan
| | - Shinichiro Okauchi
- Division of Respiratory Medicine, Mito Medical Center, University of Tsukuba-Mito Kyodo General Hospital, Mito, Japan
| | - Gen Ogara
- Division of Respiratory Medicine, Mito Medical Center, University of Tsukuba-Mito Kyodo General Hospital, Mito, Japan
| | - Tomohiro Tamura
- Respiratory Center, Ibaraki Prefectural Central Hospital, Kasama, Japan
| | - Norihiro Kikuchi
- Division of Respiratory Medicine, National Hospital Organization Kasumigaura Medical Center, Tsuchiura, Japan
| | - Kunihiko Miyazaki
- Division of Respiratory Medicine, Ryugasaki Saiseikai Hospital, Ryugasaki, Japan
| | - Shigen Hayashi
- Division of Respiratory Medicine, Ibaraki Seinan Medical Center Hospital, Sakai, Japan
| | - Takaaki Yamashita
- Division of Respiratory Medicine, JA Toride Medical Center Hospital, Toride, Japan
| | - Koichi Kurishima
- Division of Respiratory Medicine, Tsukuba Medical Center Hospital, Tsukuba, Japan
| | - Masaharu Inagaki
- Division of Thoracic Surgery, Tsuchiura Kyodo General Hospital, Tsuchiura, Japan
| | - Hiroaki Satoh
- Division of Respiratory Medicine, Mito Medical Center, University of Tsukuba-Mito Kyodo General Hospital, Mito, Japan
| | - Takayuki Kaburagi
- Respiratory Center, Ibaraki Prefectural Central Hospital, Kasama, Japan
| | - Takeo Endo
- Departments of Respiratory Medicine and Surgery, National Hospital Organization Mito Medical Center, Ibarakimachi, Japan
| | - Nobuyuki Hizawa
- Division of Respiratory Medicine, Faculty of Clinical Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
11
|
Lv L, Miao Q, Zhan S, Chen P, Liu W, Lv J, Yan W, Wang D, Liu H, Yin J, Feng J, Song Y, Ye M, Lv T. LKB1 dictates sensitivity to immunotherapy through Skp2-mediated ubiquitination of PD-L1 protein in non-small cell lung cancer. J Immunother Cancer 2024; 12:e009444. [PMID: 39694700 PMCID: PMC11660338 DOI: 10.1136/jitc-2024-009444] [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: 04/15/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Loss-of-function mutations of liver kinase B (LKB1, also termed as STK11 (serine/threonine kinase 11)) are frequently detected in patients with non-small cell lung cancer (NSCLC). The LKB1 mutant NSCLC was refractory to almost all the antitumor treatments, including programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) blockade therapy. Unfortunately, mechanisms underlying resistance to immunotherapy are not fully understood. In this study, we deciphered how LKB1 regulated sensitivity to anti-PD-1/PD-L1 immunotherapy. METHODS We investigated the mutational landscape of LKB1 mutant NSCLC in next generation sequencing (NGS) data sets. Expression of LKB1, PD-L1 and S-phase kinase-associated protein 2 (Skp2) in NSCLC samples were assessed by immunohistochemistry (IHC). The tumor microenvironment (TME) profiling of LKB1 wild type (WT) and mutant NSCLC was performed using fluorescent multiplex IHC. Mass spectrometry and enrichment analysis were used to identify LKB1 interacting proteins. Mechanistic pathways were explored by immunoblotting, ubiquitination assay, cycloheximide chase assay and immunoprecipitation assay. RESULTS By using NGS data sets and histological approaches, we demonstrated that LKB1 status was positively associated with PD-L1 protein expression and conferred a T cell-enriched "hot" TME in NSCLC. Patients with good responses to anti-PD-1/PD-L1 immunotherapy possessed a high level of LKB1 and PD-L1. Skp2 emerged as the molecular hub connecting LKB1 and PD-L1, by which Skp2 catalyzed K63-linked polyubiquitination on K136 and K280 residues to stabilize PD-L1 protein. Inhibition of Skp2 expression by short hairpin RNA or its E3 ligase activity by compound #25 abrogated intact expression of PD-L1 in vitro and generated a T cell-excluded "cold" TME in vivo. Thus, the LKB1-Skp2-PD-L1 regulatory loop was crucial for retaining PD-L1 protein expression and manipulation of this pathway would be a feasible approach for TME remodeling. CONCLUSION LKB1 and Skp2 are required for intact PD-L1 protein expression and TME remodeling in NSCLC. Inhibition of Skp2 resulted in a conversion from "hot" TME to "cold" TME and abrogated therapeutic outcomes of immunotherapy. Screening LKB1 and Skp2 status would be helpful to select recipients who may benefit from anti-PD-1/PD-L1 immunotherapy.
Collapse
MESH Headings
- Humans
- AMP-Activated Protein Kinase Kinases/analysis
- AMP-Activated Protein Kinase Kinases/genetics
- AMP-Activated Protein Kinase Kinases/metabolism
- S-Phase Kinase-Associated Proteins/analysis
- S-Phase Kinase-Associated Proteins/antagonists & inhibitors
- S-Phase Kinase-Associated Proteins/genetics
- S-Phase Kinase-Associated Proteins/metabolism
- B7-H1 Antigen/analysis
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/metabolism
- Ubiquitination
- Loss of Function Mutation
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Immunotherapy/methods
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/immunology
- Immune Checkpoint Inhibitors/pharmacology
- Immune Checkpoint Inhibitors/therapeutic use
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Gene Expression Regulation, Neoplastic/immunology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Line, Tumor
- Specific Pathogen-Free Organisms
- Mice, Inbred C57BL
- Male
- Animals
- Mice
- Xenograft Model Antitumor Assays
- Female
- Middle Aged
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Ubiquitin-Protein Ligases/analysis
- Ubiquitin-Protein Ligases/antagonists & inhibitors
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
- Cohort Studies
Collapse
Affiliation(s)
- Liting Lv
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, China
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Qing Miao
- Department of Pharmacy, the 971 Hospital of PLA Navy, Qingdao, China
| | - Sutong Zhan
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Peilin Chen
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Wei Liu
- Liaoning Kanghui Biotechnology Co. Ltd, Shenyang, China
| | - Jiawen Lv
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Wenjie Yan
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Dong Wang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Hongbing Liu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jie Yin
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jian Feng
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Mingxiang Ye
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Tangfeng Lv
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
12
|
Zhang Y, Sun D, Han W, Yang Z, Lu Y, Zhang X, Wang Y, Zhang C, Liu N, Hou H. SMARCA4 mutations and expression in lung adenocarcinoma: prognostic significance and impact on the immunotherapy response. FEBS Open Bio 2024; 14:2086-2103. [PMID: 39322625 PMCID: PMC11609588 DOI: 10.1002/2211-5463.13899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/20/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024] Open
Abstract
The switch/sucrose non-fermenting (SWI/SNF) complex family includes important chromatin-remodeling factors that are frequently mutated in lung adenocarcinoma (LUAD). However, the role of one family member, SMARCA4, in LUAD prognosis and immunotherapy sensitivity remains unclear. In the present study, 6745 LUAD samples from the cBioPortal database were used to analyze the relationships between SMARCA4 mutations and patient prognoses and clinical characteristics. Additionally, we examined the correlation between SMARCA4 mutations and prognosis in patients treated with immunotherapy using two immune-related datasets. SMARCA4 mutations and low expression were associated with shorter survival, and mutations were associated with a high tumor mutational burden and high microsatellite instability. SMARCA4 mutations were accompanied by KRAS, KEAP1, TP53 and STK11 mutations. No significant difference was observed in the immunotherapy response between patients with and without SMARCA4 mutations. When KRAS or STK11 mutations were present, immunotherapy effectiveness was poorer; however, when both SMARCA4 and TP53 mutations were present, immunotherapy was more effective. Furthermore, low SMARCA4 expression predicted a higher immunophenoscore, and SMARCA4 expression was correlated with certain immune microenvironment features. Taken together, our results suggest that SMARCA4 mutations and low expression might be associated with poor LUAD prognosis. Additionally, immunotherapy efficacy in patients with SMARCA4 mutations depended on the co-mutant genes. Thus, SMARCA4 could be an important factor to be considered for LUAD diagnosis and treatment.
Collapse
Affiliation(s)
- Yuming Zhang
- Precision Medicine Center of OncologyThe Affiliated Hospital of Qingdao University, Qingdao UniversityChina
| | - Dantong Sun
- Department of Medical OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Weizhong Han
- Department of Respiratory MedicineThe Affiliated Hospital of Qingdao UniversityChina
| | - Zhen Yang
- Department of PathologyThe Affiliated Hospital of Qingdao University, Qingdao UniversityChina
| | - Yongzhi Lu
- Department of OncologyQingdao Municipal HospitalChina
| | - Xuchen Zhang
- Precision Medicine Center of OncologyThe Affiliated Hospital of Qingdao University, Qingdao UniversityChina
| | - Yongjie Wang
- Department of Thoracic SurgeryThe Affiliation Hospital of Qingdao UniversityChina
| | - Chuantao Zhang
- Department of OncologyThe Affiliated Hospital of Qingdao UniversityChina
| | - Ning Liu
- Department of OncologyThe Affiliated Hospital of Qingdao UniversityChina
| | - Helei Hou
- Department of OncologyThe Affiliated Hospital of Qingdao UniversityChina
| |
Collapse
|
13
|
Liu H, Hong Q, Zheng S, Zhang M, Cai L. Effective treatment strategies and key factors influencing therapeutic efficacy in advanced SMARCA4-deficient non-small cell lung cancer. Lung Cancer 2024; 198:108022. [PMID: 39541774 DOI: 10.1016/j.lungcan.2024.108022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION SMARCA4/BRG1-deficient non-small cell lung cancer (SD-NSCLC) with high invasiveness and poor prognosis is associated with primary resistance to standard treatment, especially in late-stage patients. This study aimed to explore effective treatments and identify critical factors impacting therapeutic efficacy to enhance outcomes for SD-NSCLC patients. METHODS 103 SD-NSCLC patients in stage III/IV diagnosed by immunohistochemistry from May 2019 to March 2024 were included in this study. We assessed the patients' clinical and genetic features, analyzed the clinical outcomes of local treatment and immunotherapy according to the TNM stage, and further evaluated the factors impacting therapeutic efficacy. RESULTS In stage III patients, no significant differences in the median progression-free survival (mPFS) and median overall survival (mOS) were observed between patients receiving local treatment at the primary site and those who did not (p > 0.05), while adding ICIs (immune checkpoint inhibitors) to local treatment significantly improved mPFS compared with non-ICIs (15.0 vs. 7.7 months, p = 0.033), though not mOS (p > 0.05). For stage IV patients, ICIs significantly improved mPFS (8.9 vs. 4.2 months, p = 0.006) and mOS (19.7 vs. 13.1 months, p = 0.007) compared to non-ICIs treatments. However, among ICIs-treated patients, the addition of local treatment to the primary lesion did not significantly affect mPFS and mOS (p > 0.05). Patients with STK11/KEAP1 mutations had significantly shorter mPFS (3.6 vs. 16.2 months, p = 0.001) and mOS (17.7 vs. 31.3 months, p = 0.002), while no significant difference was observed in mPFS and mOS in patients with different tumor mutation burden (TMB) and PD-L1 expression levels. CONCLUSION The addition of ICIs to local treatment shows promising results for locally advanced patients with SD-NSCLC, and first-line ICIs are associated with improved survival in metastatic SD-NSCLC. STK11/KEAP1 mutations may be linked to reduced efficacy of immunotherapy.
Collapse
Affiliation(s)
- Hui Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China
| | - Qiyuan Hong
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China
| | - Shuohan Zheng
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China
| | - Meifang Zhang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China; Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, PR China
| | - Ling Cai
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China; State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China.
| |
Collapse
|
14
|
Tenekeci AK, Unal AA, Ceylan F, Nahit Sendur MA. An updated overview of K-RAS G12C inhibitors in advanced stage non-small cell lung cancer. Future Oncol 2024; 20:3019-3038. [PMID: 39360933 PMCID: PMC11572139 DOI: 10.1080/14796694.2024.2407280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 09/18/2024] [Indexed: 11/16/2024] Open
Abstract
The discovery of KRAS mutations, particularly the KRASG12C variant, has been a milestone in understanding the molecular underpinnings of non-small cell lung cancer (NSCLC). These mutations are associated with aggressive tumor behavior and resistance to conventional therapies, highlighting the urgent need for targeted interventions. In this comprehensive review, we analyze the advancements in KRAS G12C inhibitors for the treatment of non-small cell lung cancer. Literature search is made from PubMed, Medline ASCO and ESMO Annual Meetings abstracts by using the following search keywords: "sotorasib", "adagrasib", "divarasib" and "KRAS G12C inhibitors." The last search was on 5 June 2024. This review highlights the importance of pharmacokinetics, pharmacodynamics and potential adverse effects for treating individual patients and ensuring the best outcomes. Additionally, the review discusses research identifying biomarkers that can predict therapy responses and mentions the combination strategies to overcome resistance. Results of the studies and ongoing clinical trials are also briefly summarized in this review. KRASG12C inhibitors sotorasib, adagrasib and the newer divarasib, has revolutionized treating patients harboring this mutation. Ongoing studies and future clinical trials will refine our understandings with the ultimate goal of improving survival and quality of life for patients with this challenging disease.
Collapse
Affiliation(s)
| | | | - Furkan Ceylan
- Ankara Bilkent City Hospital, Department of Medical Oncology, Ankara, Turkey
| | - Mehmet Ali Nahit Sendur
- Ankara Yildirim Beyazit University Faculty of Medicine and Ankara Bilkent City Hospital, Department of Medical Oncology, Ankara, Turkey
| |
Collapse
|
15
|
Pavelescu LA, Enache RM, Roşu OA, Profir M, Creţoiu SM, Gaspar BS. Predictive Biomarkers and Resistance Mechanisms of Checkpoint Inhibitors in Malignant Solid Tumors. Int J Mol Sci 2024; 25:9659. [PMID: 39273605 PMCID: PMC11395316 DOI: 10.3390/ijms25179659] [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: 08/22/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Predictive biomarkers for immune checkpoint inhibitors (ICIs) in solid tumors such as melanoma, hepatocellular carcinoma (HCC), colorectal cancer (CRC), non-small cell lung cancer (NSCLC), endometrial carcinoma, renal cell carcinoma (RCC), or urothelial carcinoma (UC) include programmed cell death ligand 1 (PD-L1) expression, tumor mutational burden (TMB), defective deoxyribonucleic acid (DNA) mismatch repair (dMMR), microsatellite instability (MSI), and the tumor microenvironment (TME). Over the past decade, several types of ICIs, including cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors, anti-programmed cell death 1 (PD-1) antibodies, anti-programmed cell death ligand 1 (PD-L1) antibodies, and anti-lymphocyte activation gene-3 (LAG-3) antibodies have been studied and approved by the Food and Drug Administration (FDA), with ongoing research on others. Recent studies highlight the critical role of the gut microbiome in influencing a positive therapeutic response to ICIs, emphasizing the importance of modeling factors that can maintain a healthy microbiome. However, resistance mechanisms can emerge, such as increased expression of alternative immune checkpoints, T-cell immunoglobulin (Ig), mucin domain-containing protein 3 (TIM-3), LAG-3, impaired antigen presentation, and alterations in the TME. This review aims to synthesize the data regarding the interactions between microbiota and immunotherapy (IT). Understanding these mechanisms is essential for optimizing ICI therapy and developing effective combination strategies.
Collapse
Affiliation(s)
- Luciana Alexandra Pavelescu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Robert Mihai Enache
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Oana Alexandra Roşu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Monica Profir
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Sanda Maria Creţoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Bogdan Severus Gaspar
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Surgery Clinic, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| |
Collapse
|
16
|
Li K, Wu L, Wang H, Fu Z, Gao J, Liu X, Fan Y, Qin X, Ni D, Wang J, Xie D. Apoptosis and cuproptosis Co-activated Copper-based metal-organic frameworks for cancer therapy. J Nanobiotechnology 2024; 22:546. [PMID: 39237931 PMCID: PMC11378619 DOI: 10.1186/s12951-024-02828-3] [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: 06/21/2024] [Accepted: 09/01/2024] [Indexed: 09/07/2024] Open
Abstract
Lung cancer, predominantly non-small cell lung cancer (NSCLC), remains a significant global health challenge, with limited therapeutic options for patients with KRAS-mutated tumors. Herein, a copper-based metal-organic framework (Cu-MOF) was applied as a novel cuproptosis-mediated nanoplatform for lung cancer therapy. Cu-MOF would disassemble and liberate copper ions under the acidic microenvironment of lysosomes of cancer cells, initiating a cascade of cellular events. The released copper ions catalyzes the Fenton reaction, generating hydroxyl radicals that induce oxidative damage, leading to cytoskeletal disruption and activation of caspase-3, ultimately triggering apoptosis. Simultaneously, with the mediation of the key regulatory factor FDX1, we found that the copper ions binding to the mitochondrial protein DLAT could result in the loss of iron-sulfur cluster proteins and aggregation of lipoylated proteins, which culminated in proteotoxic stress-induced cuproptosis. The pronounced anti-tumor effects of Cu-MOF with apoptosis and cuproptosis were confirmed both in vitro and in vivo experiments. Such dual induction of apoptosis and cuproptosis by Cu-MOF presents a promising therapeutic strategy for NSCLC, particularly for KRAS-mutated tumors, and expands potential applications of Cu-based nanomateirals for other cancers.
Collapse
Affiliation(s)
- Kun Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Leilei Wu
- Department of Thoracic Surgery, Hangzhou Institute of Medicine (HIM), Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, 310005, P. R. China
| | - Han Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Zi Fu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Jiani Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Xiucheng Liu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Yongfei Fan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Xichun Qin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Dalong Ni
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China.
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215163, P. R. China.
| | - Jing Wang
- Department of Radiology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, P. R. China.
| | - Dong Xie
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China.
| |
Collapse
|
17
|
Schoenfeld AJ, Lee SM, Doger de Spéville B, Gettinger SN, Häfliger S, Sukari A, Papa S, Rodríguez-Moreno JF, Graf Finckenstein F, Fiaz R, Catlett M, Chen G, Qi R, Masteller EL, Gontcharova V, He K. Lifileucel, an Autologous Tumor-Infiltrating Lymphocyte Monotherapy, in Patients with Advanced Non-Small Cell Lung Cancer Resistant to Immune Checkpoint Inhibitors. Cancer Discov 2024; 14:1389-1402. [PMID: 38563600 PMCID: PMC11294887 DOI: 10.1158/2159-8290.cd-23-1334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/18/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
In this phase 2 multicenter study, we evaluated the efficacy and safety of lifileucel (LN-145), an autologous tumor-infiltrating lymphocyte cell therapy, in patients with metastatic non-small cell lung cancer (mNSCLC) who had received prior immunotherapy and progressed on their most recent therapy. The median number of prior systemic therapies was 2 (range, 1-6). Lifileucel was successfully manufactured using tumor tissue from different anatomic sites, predominantly lung. The objective response rate was 21.4% (6/28). Responses occurred in tumors with profiles typically resistant to immunotherapy, such as PD-L1-negative, low tumor mutational burden, and STK11 mutation. Two responses were ongoing at the time of data cutoff, including one complete metabolic response in a PD-L1-negative tumor. Adverse events were generally as expected and manageable. Two patients died of treatment-emergent adverse events: cardiac failure and multiple organ failure. Lifileucel is a potential treatment option for patients with mNSCLC refractory to prior therapy. Significance: Autologous tumor-infiltrating lymphocyte therapy lifileucel was administered to 28 patients with heavily pretreated metastatic non-small cell lung cancer (mNSCLC). Responses were observed in patients with driver mutations, and various tumor mutational burdens and PD-L1 expression, potentially addressing an unmet medical need in patients with mNSCLC refractory to prior therapy. See related commentary by Lotze et al., p. 1366.
Collapse
Affiliation(s)
- Adam J. Schoenfeld
- Division of Solid Tumor Oncology, Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Sylvia M. Lee
- Clinical Research Division, Department of Medicine, Fred Hutchinson Cancer Center, Seattle, Washington.
| | | | - Scott N. Gettinger
- Division of Medical Oncology, Department of Medicine, Yale Cancer Center, North Haven, Connecticut.
| | - Simon Häfliger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Ammar Sukari
- Department of Oncology, Barbara Ann Karmanos Cancer Hospital, Detroit, Michigan.
| | - Sophie Papa
- School of Cancer and Pharmaceutical Sciences, King’s College London, London, United Kingdom.
- Department of Medical Oncology, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom.
| | - Juan F. Rodríguez-Moreno
- Department of Medical Oncology, Hospital Universitario HM Sanchinarro, Centro Integral Oncológico Clara Campal, Madrid, Spain.
| | | | - Rana Fiaz
- Iovance Biotherapeutics, Inc., San Carlos, California.
| | | | - Guang Chen
- Iovance Biotherapeutics, Inc., San Carlos, California.
| | - Rongsu Qi
- Iovance Biotherapeutics, Inc., San Carlos, California.
| | | | | | - Kai He
- Division of Medical Oncology, Department of Internal Medicine, Thoracic Oncology Program, Ohio State University, Columbus, Ohio.
| |
Collapse
|
18
|
Lan T, Arastu S, Lam J, Kim H, Wang W, Wang S, Bhatt V, Lopes EC, Hu Z, Sun M, Luo X, Ghergurovich JM, Su X, Rabinowitz JD, White E, Guo JY. Glucose-6-phosphate dehydrogenase maintains redox homeostasis and biosynthesis in LKB1-deficient KRAS-driven lung cancer. Nat Commun 2024; 15:5857. [PMID: 38997257 PMCID: PMC11245543 DOI: 10.1038/s41467-024-50157-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
Cancer cells depend on nicotinamide adenine dinucleotide phosphate (NADPH) to combat oxidative stress and support reductive biosynthesis. One major NADPH production route is the oxidative pentose phosphate pathway (committed step: glucose-6-phosphate dehydrogenase, G6PD). Alternatives exist and can compensate in some tumors. Here, using genetically-engineered lung cancer mouse models, we show that G6PD ablation significantly suppresses KrasG12D/+;Lkb1-/- (KL) but not KrasG12D/+;P53-/- (KP) lung tumorigenesis. In vivo isotope tracing and metabolomics reveal that G6PD ablation significantly impairs NADPH generation, redox balance, and de novo lipogenesis in KL but not KP lung tumors. Mechanistically, in KL tumors, G6PD ablation activates p53, suppressing tumor growth. As tumors progress, G6PD-deficient KL tumors increase an alternative NADPH source from serine-driven one carbon metabolism, rendering associated tumor-derived cell lines sensitive to serine/glycine depletion. Thus, oncogenic driver mutations determine lung cancer dependence on G6PD, whose targeting is a potential therapeutic strategy for tumors harboring KRAS and LKB1 co-mutations.
Collapse
Affiliation(s)
- Taijin Lan
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | - Sara Arastu
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | - Jarrick Lam
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | - Hyungsin Kim
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | - Wenping Wang
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | - Samuel Wang
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | | | - Eduardo Cararo Lopes
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, 08854, USA
| | - Zhixian Hu
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | - Michael Sun
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | - Xuefei Luo
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
| | | | - Xiaoyang Su
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Joshua D Rabinowitz
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
- Department of Chemistry, Princeton University, Princeton, NJ, 08544, USA
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA
| | - Eileen White
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ, 08854, USA
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ, 08544, USA
| | - Jessie Yanxiang Guo
- Rutgers Cancer Institute, New Brunswick, NJ, 08901, USA.
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
- Department of Chemical Biology, Rutgers Ernest Mario School of Pharmacy, Piscataway, NJ, 08854, USA.
| |
Collapse
|
19
|
Frost N, Reck M. Non-Small Cell Lung Cancer Metastatic Without Oncogenic Alterations. Am Soc Clin Oncol Educ Book 2024; 44:e432524. [PMID: 38669613 DOI: 10.1200/edbk_432524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
This overview provides a thorough review of current treatment approaches for first-line management of nononcogenic addicted non-small cell lung cancer. We also address pertinent clinical decision-making queries encountered in everyday practice, such as the optimal treatment strategy for PD-L1-high patients, predictive factors for response to immune checkpoint inhibitors (ICI) both in terms of patient and cancer characteristics, the potential benefits of dual checkpoint blockade, and the unresolved issue of safe discontinuation strategies for long-term responders. Around one in five patients falls into this latter category while the majority develop either primary or acquired resistance to ICI-based first-line therapy, necessitating effective subsequent lines of treatment. Docetaxel, with or without combination of antiangiogenic agents, serves as the backbone of treatment, although evidence in the post-ICI setting is limited. Given that an inflamed tumor microenvironment (TME) is crucial for ICI responses, targeting the TME in cases of acquired resistance alongside continued ICI administration appears rational, although clinical trials so far have failed to confirm this hypothesis. Antibody-drug conjugates have emerged as a promising treatment modality, offering the potential for reduced toxicity and improved efficacy by targeting specific cancer antigens. Moreover, several chemotherapy-free approaches are currently under investigation for treatment-naïve patients, including alternative ICI and drugs targeting epitopes on both cancer and immune cells.
Collapse
Affiliation(s)
- Nikolaj Frost
- Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Infectious Diseases and Pulmonary Medicine, Berlin, Germany
| | - Martin Reck
- Department of Thoracic Oncology, Airway Research Center North, German Center for Lung Research, LungenClinic, Grosshansdorf, Germany
| |
Collapse
|
20
|
Li T, Liu J, Zhou Y, Huang S, Wang D, Chen J, Fu Y, He P. Clinical relevance of somatic mutations in Chinese lung adenocarcinoma and their prognostic implications for survival. Cancer Med 2024; 13:e7227. [PMID: 38770632 PMCID: PMC11106684 DOI: 10.1002/cam4.7227] [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: 09/11/2023] [Revised: 04/07/2024] [Accepted: 04/18/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND To comprehensively elucidate the genomic and mutational features of lung cancer cases, and lung adenocarcinoma (LUAD), it is imperative to conduct ongoing investigations into the genomic landscape. In this study, we aim to analyze the somatic mutation profile and assessed the significance of these informative genes utilizing a retrospective LUAD cohort. METHODS A total of 247 Chinese samples were analyzed to exhibit the tumor somatic genomic alterations in patients with LUAD. The Cox regression analysis was employed to identify prognosis-related genes and establish a predictive model for stratifying patients with LUAD. RESULTS In the Dianjiang People's Hospital (DPH) cohort, the top five frequent mutated genes were (Epidermal growth factor receptor) EGFR (68%), TP53 (30%), RBM10 (13%), LRP1B (9%), and KRAS (9%). Of which, EGFR is a mostly altered driver gene, and most mutation sites are located in tyrosine kinase regions. Oncogene pathway alteration and mutation signature analysis demonstrated the RTK-RAS pathway alteration, and smoking was the main carcinogenic factor of the DPH cohort. Furthermore, we identified 34 driver genes in the DPH cohort, including EGFR (68%), TP53 (30.4%), RBM10 (12.6%), KRAS (8.5%), LRP1B (8.5%), and so on, and 45 Clinical Characteristic-Related Genes (CCRGs) were found to closely related to the clinical high-risk factors. We developed a Multiple Parameter Gene Mutation (MPGM) risk model by integrating critical genes and oncogenic pathway alterations in LUAD patients from the DPH cohort. Based on publicly available LUAD datasets, we identified five genes, including BRCA2, Anaplastic lymphoma kinase (ALK), BRAF, EGFR, and Platelet-Derived Growth Factor Receptor Alpha (PDGFRA), according to the multivariable Cox regression analysis. The MPGM-low group showed significantly better overall survival (OS) compared to the MPGM-high group (p < 0.0001, area under the curve (AUC) = 0.754). The robust performance was validated in 55 LUAD patients from the DPH cohort and another LUAD dataset. Immune characteristics analysis revealed a higher proportion of primarily DCs and mononuclear cells in the MPGM-low risk group, while the MPGM-high risk group showed lower immune cells and higher tumor cell infiltration. CONCLUSION This study provides a comprehensive genomic landscape of Chinese LUAD patients and develops an MPGM risk model for LUAD prognosis stratification. Further follow-up will be performed for the patients in the DPH cohort consistently to explore the resistance and prognosis genetic features.
Collapse
Affiliation(s)
- Tongxin Li
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of ChongqingChongqingChina
| | - Jie Liu
- Department of Thoracic Surgery, Southwest HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Yu Zhou
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of ChongqingChongqingChina
| | - Shengyuan Huang
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of ChongqingChongqingChina
| | - Dong Wang
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of ChongqingChongqingChina
| | - Jianrong Chen
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of ChongqingChongqingChina
| | - Yong Fu
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of ChongqingChongqingChina
| | - Ping He
- Department of Cardiac Surgery, Southwest HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| |
Collapse
|
21
|
Bischoff P, Reck M, Overbeck T, Christopoulos P, Rittmeyer A, Lüders H, Kollmeier J, Kulhavy J, Kemper M, Reinmuth N, Röper J, Janning M, Sommer L, Aguinarte L, Koch M, Wiesweg M, Wesseler C, Waller CF, Kauffmann-Guerrero D, Stenzinger A, Stephan-Falkenau S, Trautmann M, Lassmann S, Tiemann M, Klauschen F, Sebastian M, Griesinger F, Wolf J, Loges S, Frost N. Outcome of First-Line Treatment With Pembrolizumab According to KRAS/TP53 Mutational Status for Nonsquamous Programmed Death-Ligand 1-High (≥50%) NSCLC in the German National Network Genomic Medicine Lung Cancer. J Thorac Oncol 2024; 19:803-817. [PMID: 38096950 DOI: 10.1016/j.jtho.2023.12.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 01/18/2024]
Abstract
INTRODUCTION Programmed death-ligand 1 expression currently represents the only validated predictive biomarker for immune checkpoint inhibition in metastatic NSCLC in the clinical routine, but it has limited value in distinguishing responses. Assessment of KRAS and TP53 mutations (mut) as surrogate for an immunosupportive tumor microenvironment (TME) might help to close this gap. METHODS A total of 696 consecutive patients with programmed death-ligand 1-high (≥50%), nonsquamous NSCLC, having received molecular testing within the German National Network Genomic Medicine Lung Cancer between 2017 and 2020, with Eastern Cooperative Oncology Group performance status less than or equal to 1 and pembrolizumab as first-line palliative treatment, were included into this retrospective cohort analysis. Treatment efficacy and outcome according to KRAS/TP53 status were correlated with TME composition and gene expression analysis of The Cancer Genome Atlas lung adenocarcinoma cohort. RESULTS Proportion of KRASmut and TP53mut was 53% (G12C 25%, non-G12C 28%) and 51%, respectively. In KRASmut patients, TP53 comutations increased response rates (G12C: 69.7% versus 46.5% [TP53mut versus wild-type (wt)], p = 0.004; non-G12C: 55.4% versus 39.5%, p = 0.03), progression-free survival (G12C: hazard ratio [HR] = 0.59, p = 0.009, non-G12C: HR = 0.7, p = 0.047), and overall survival (G12C: HR = 0.72, p = 0.16, non-G12C: HR = 0.56, p = 0.002), whereas no differences were observed in KRASwt patients. After a median follow-up of 41 months, G12C/TP53mut patients experienced the longest progression-free survival and overall survival (33.7 and 65.3 mo), which correlated with high tumor-infiltrating lymphocyte densities in the TME and up-regulation of interferon gamma target genes. Proinflammatory pathways according to TP53 status (mut versus wt) were less enhanced and not different in non-G12C and KRASwt, respectively. CONCLUSIONS G12C/TP53 comutations identify a subset of patients with a very favorable long-term survival with immune checkpoint inhibitor monotherapy, mediated by highly active interferon gamma signaling in a proinflammatory TME.
Collapse
Affiliation(s)
- Philip Bischoff
- Institute of Pathology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany; BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Berlin, Germany; German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Reck
- Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany
| | - Tobias Overbeck
- Department of Haematology and Medical Oncology, University Medical Center Göttingen and Lungentumorzentrum Universität Göttingen, Göttingen, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Achim Rittmeyer
- Department of Thoracic Oncology, LKI Lungenfachklinik Immenhausen, Immenhausen, Germany
| | - Heike Lüders
- Klinik für Pneumologie-Evangelische Lungenklinik Berlin Buch, Berlin, Germany
| | - Jens Kollmeier
- Helios Klinikum Emil von Behring, Lungenklinik Heckeshorn, Berlin, Germany; Berlin Lung Institute, Berlin, Germany
| | - Jonas Kulhavy
- Translational Oncology/Early Clinical Trial Unit (ECTU), Comprehensive Cancer Center Mainfranken and Bavarian Cancer Research Center (BZKF), University Hospital Wuerzburg, Wuerzburg, Germany
| | - Marcel Kemper
- Department of Medicine A for Hematology, Oncology and Pneumology, University Hospital Muenster, Muenster, Germany
| | - Niels Reinmuth
- Asklepios Lung Clinic, member of the German Center for Lung Research (DZL), Munich-Gauting, Germany
| | - Julia Röper
- Department of Hematology and Oncology, Pius-Hospital, University Dept. of Internal Medicine-Oncology, Oldenburg, Germany
| | - Melanie Janning
- DKFZ-Hector Cancer Institute and Department of Personalized Oncology at the University Hospital Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Linna Sommer
- Department of Thoracic Oncology, Carl-Gustav-Carus Dresden University Hospital, Dresden, Germany
| | - Lukas Aguinarte
- Hematology/Oncology, Department of Medicine II, University Hospital Frankfurt, Frankfurt, Germany
| | - Myriam Koch
- University Hospital Regensburg, Department of Internal Medicine 2, Regensburg, Germany
| | - Marcel Wiesweg
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Claas Wesseler
- Department of Thoracic Oncology, Asklepios Klinikum Harburg, Hamburg, Germany
| | - Cornelius F Waller
- Department of Haematology, Oncology and Stem Cell Transplantation, University Medical Centre Freiburg and Faculty of Medicine, Freiburg, Germany
| | - Diego Kauffmann-Guerrero
- Department of Medicine V, University Hospital, LMU Munich, Member of the German Center for Lung Research (DZL-CPCM), Munich, Germany
| | | | | | - Marcel Trautmann
- University of Münster, Division of Translational Pathology, Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Silke Lassmann
- Institute for Surgical Pathology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Frederick Klauschen
- Institute of Pathology, Ludwig-Maximilians-University, Munich, Germany; Berlin Institute for the Foundation of Learning and Data (BIFOLD) and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Martin Sebastian
- Hematology/Oncology, Department of Medicine II, University Hospital Frankfurt, Frankfurt, Germany
| | - Frank Griesinger
- Department of Hematology and Oncology, Pius-Hospital, University Dept. of Internal Medicine-Oncology, Oldenburg, Germany
| | - Jürgen Wolf
- Department I of Internal Medicine, Center for Integrated Oncology (CIO), University Hospital of Cologne, Cologne, Germany
| | - Sonja Loges
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Nikolaj Frost
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin (Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health), Berlin, Germany.
| |
Collapse
|
22
|
Zheng J, Deng Y, Huang B, Chen X. Prognostic implications of STK11 with different mutation status and its relationship with tumor-infiltrating immune cells in non-small cell lung cancer. Front Immunol 2024; 15:1387896. [PMID: 38736875 PMCID: PMC11082287 DOI: 10.3389/fimmu.2024.1387896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
Background Mutations in STK11 (STK11Mut) gene may present a negative impact on survival in Non-small Cell Lung Cancer (NSCLC) patients, however, its relationship with immune related genes remains unclear. This study is to unveil whether overexpressed- and mutated-STK11 impact survival in NSCLC and to explore whether immune related genes (IRGs) are involved in STK11 mutations. Methods 188 NSCLC patients with intact formalin-fixed paraffin-embedded (FFPE) tissue available for detecting STK11 protein expression were included in the analysis. After immunohistochemical detection of STK11 protein, patients were divided into high STK11 expression group (STK11High) and low STK11 expression group (STK11Low), and then Kaplan-Meier survival analysis and COX proportional hazards model were used to compare the overall survival (OS) and progression-free survival (PFS) of the two groups of patients. In addition, the mutation data from the TCGA database was used to categorize the NSCLC population, namely STK11 Mutated (STK11Mut) and wild-type (STK11Wt) subgroups. The difference in OS between STK11Mut and STK11Wt was compared. Finally, bioinformatics analysis was used to compare the differences in IRGs expression between STK11Mut and STK11Wt populations. Results The median follow-up time was 51.0 months (range 3.0 - 120.0 months) for real-life cohort. At the end of follow-up, 64.36% (121/188) of patients experienced recurrence or metastasis. 64.89% (122/188) of patients ended up in cancer-related death. High expression of STK11 was a significant protective factor for NSCLC patients, both in terms of PFS [HR=0.42, 95% CI= (0.29-0.61), P<0.001] and OS [HR=0.36, 95% CI= (0.25, 0.53), P<0.001], which was consistent with the finding in TCGA cohorts [HR=0.76, 95%CI= (0.65, 0.88), P<0.001 HR=0.76, 95%CI= (0.65, 0.88), P<0.001]. In TCGA cohort, STK11 mutation was a significant risk factor for NSCLC in both lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD) histology in terms of OS [HR=6.81, 95%CI= (2.16, 21.53), P<0.001; HR=1.50, 95%CI= (1.00, 2.26), P=0.051, respectively]. Furthermore, 7 IRGs, namely CALCA, BMP6, S100P, THPO, CGA, PCSK1 and MUC5AC, were found significantly overexpressed in STK11-mutated NSCLC in both LUSC and LUAD histology. Conclusions Low STK11 expression at protein level and presence of STK11 mutation were associated with poor prognosis in NSCLC, and mutated STK11 might probably alter the expression IRGs profiling.
Collapse
Affiliation(s)
- Jianqing Zheng
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Yujie Deng
- Department of Medical Oncology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Bifen Huang
- Department of Obstetrics and Gynecology, Quanzhou Medical College People’s Hospital Affiliated, Quanzhou, Fujian, China
| | - Xiaohui Chen
- Department of Thoracic Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- The Graduate School of Fujian Medical University, Fuzhou, Fujian, China
- Interdisciplinary Institute of Medical Engineering of Fuzhou University, Fuzhou, Fujian, China
| |
Collapse
|
23
|
Caballé-Perez E, Hernández-Pedro N, Ramos-Ramírez M, Barrios-Bernal P, Romero-Núñez E, Lucio-Lozada J, Ávila-Ríos S, Reyes-Terán G, Cardona AF, Arrieta O. Impact of KRAS G12D subtype and concurrent pathogenic mutations on advanced non-small cell lung cancer outcomes. Clin Transl Oncol 2024; 26:836-850. [PMID: 37490263 PMCID: PMC10981588 DOI: 10.1007/s12094-023-03279-2] [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: 06/26/2023] [Accepted: 07/09/2023] [Indexed: 07/26/2023]
Abstract
PURPOSE Mutations in the Kirsten rat sarcoma viral (KRAS) oncogene constitute a significant driver of lung adenocarcinoma, present in 10-40% of patients, which exhibit heterogeneous clinical outcomes, mainly driven by concurrent genetic alterations. However, characterization of KRAS mutational subtypes and their impact on clinical outcomes in Latin America is limited. METHODS A cohort study was conducted at the National Cancer Institute (INCan) of Mexico. Individuals with advance-staged of adenocarcinoma and KRAS mutations, detected by next-generation sequencing, having undergone at least one line of therapy were included for analysis. Clinical and pathological characteristics were retrieved from institutional database from June 2014 to March 2023. RESULTS KRAS was identified in fifty-four (15.6%) of 346 patients, among which 50 cases were included for analysis. KRASG12D (n = 16, 32%) and KRASG12C (n = 16, 32%) represented the most prevalent subtypes. KRASG12D mutations were associated with female (p = 0.018), never smokers (p = 0.108), and concurrences with EGFR (25.0% vs. 17.6%, p = 0.124) and CDKN2A (18.8% vs. 14.7%, p = 0.157). KRASG12D patients showed a better ORR (66.6% vs. 30.0%; OR 4.66, 95% CI 1.23-17.60, p = 0.023) and on multivariate analysis was significantly associated with better PFS (HR 0.36, 95% CI 0.16-0.80; p = 0.012) and OS (HR 0.24, 95% CI 0.08-0.70; p = 0.009). CONCLUSIONS To our knowledge, this study represents the first effort to comprehensively characterize the molecular heterogeneity of KRAS-mutant NSCLC in Latin American patients. Our data reinforce the current view that KRAS-mutated NSCLC is not a single oncogene-driven disease and emphasizes the prognostic impact of diverse molecular profiles in this genomically defined subset of NSCLC. Further validation is warranted in larger multicenter Latin American cohorts to confirm our findings.
Collapse
Affiliation(s)
- Enrique Caballé-Perez
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Norma Hernández-Pedro
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Maritza Ramos-Ramírez
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Pedro Barrios-Bernal
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Eunice Romero-Núñez
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - José Lucio-Lozada
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | | | | | - Andrés F Cardona
- Thoracic Oncology Unit and Direction of Research, Science and Education, Luis Carlos Sarmiento Angulo, Cancer Treatment and Research Center (CTIC), Bogotá, Colombia
- Clinical and Translational Oncology Group, Clínica del Country, Bogotá, Colombia
- Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Oscar Arrieta
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico.
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico.
| |
Collapse
|
24
|
Zhou P, Fu Y, Tang Y, Jiang L, Wang W. Thoracic SMARCA4-deficient undifferentiated tumor: A clinicopathological and prognostic analysis of 35 cases and immunotherapy efficacy. Lung Cancer 2024; 189:107471. [PMID: 38306886 DOI: 10.1016/j.lungcan.2024.107471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/04/2024]
Abstract
BACKGROUND Thoracic SMARCA4-deficient undifferentiated tumor (SMARCA4-UT) is a recently recognized distinct clinicopathological entity according to the fifth edition of the 2021 World Health Organization Classification (WHO) for thoracic tumors. Thoracic SMARCA4-UTs are diagnostically challenging to diagnose, especially on small biopsies. METHODS We identified 35 thoracic SMARCA4-UTs from the Department of Pathology of West China Hospital, Sichuan University, between January 2017 and December 2022. In the present study, we summarized the clinicopathological features, prognostic significance and immunotherapy efficacy of thoracic SMARCA4-UTs. RESULTS All 35 patients were male, and 88.6 % were smokers. The left upper lobe (25.7 %) and mediastinum (20.0 %) were the most affected sites. 17.1 % of the patients received surgical treatment. 30.4 % of the patients were stage III, and 69.6 % were stage IV. Solid architecture (100 %), rhabdoid morphology (51.4 %) and necrosis (42.9 %) were the common histological features. Immunohistochemical staining revealed CD34 and synaptophysin positivity in most patients (76.9 % and 65.2 %, respectively). Patients had unfavorable outcomes. Patients who received immunotherapy had better OS and PFS than those who did not (p = 0.007 and p = 0.02, respectively). Five patients were evaluated for immunotherapy efficacy, and four of those patients were negative expression of PD-L1. Cases 1-4 presented TIL counts ranging from 20 to 1000/HPF. Case 5 presented TIL counts of 5-10/HPF. Mutations in SMARCA4 were confirmed in cases 4 and 5, and the TMB was 5.98 and 5.03 mutations/Mb, respectively. Case 1 achieved a CR, cases 2-4 achieved a PR, and case 5 had a PD. Five patients who received immunotherapy were all alive, with OS ranging from 10.7 to 33.6 months. CONCLUSIONS Thoracic SMARCA4-UTs exhibited an aggressive clinical course, presented solid architecture with or without necrosis and/or rhabdoid morphology, and frequently expressed CD34 and synaptophysin. Some thoracic SMARCA4-UTs appear to be associated with responsiveness to immunotherapy, suggesting the need for validation in larger series.
Collapse
Affiliation(s)
- Ping Zhou
- Department of Pathology, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Yiyun Fu
- Department of Pathology, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Yuan Tang
- Department of Pathology, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Lili Jiang
- Department of Pathology, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Weiya Wang
- Department of Pathology, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China.
| |
Collapse
|
25
|
Kazandjian S, Rousselle E, Dankner M, Cescon DW, Spreafico A, Ma K, Kavan P, Batist G, Rose AAN. The Clinical, Genomic, and Transcriptomic Landscape of BRAF Mutant Cancers. Cancers (Basel) 2024; 16:445. [PMID: 38275886 PMCID: PMC10814895 DOI: 10.3390/cancers16020445] [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: 11/30/2023] [Revised: 01/08/2024] [Accepted: 01/13/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND BRAF mutations are classified into four molecularly distinct groups, and Class 1 (V600) mutant tumors are treated with targeted therapies. Effective treatment has not been established for Class 2/3 or BRAF Fusions. We investigated whether BRAF mutation class differed according to clinical, genomic, and transcriptomic variables in cancer patients. METHODS Using the AACR GENIE (v.12) cancer database, the distribution of BRAF mutation class in adult cancer patients was analyzed according to sex, age, primary race, and tumor type. Genomic alteration data and transcriptomic analysis was performed using The Cancer Genome Atlas. RESULTS BRAF mutations were identified in 9515 (6.2%) samples among 153,834, with melanoma (31%), CRC (20.7%), and NSCLC (13.9%) being the most frequent cancer types. Class 1 harbored co-mutations outside of the MAPK pathway (TERT, RFN43) vs. Class 2/3 mutations (RAS, NF1). Across all tumor types, Class 2/3 were enriched for alterations in genes involved in UV response and WNT/β-catenin. Pathway analysis revealed enrichment of WNT/β-catenin and Hedgehog signaling in non-V600 mutated CRC. Males had a higher proportion of Class 3 mutations vs. females (17.4% vs. 12.3% q = 0.003). Non-V600 mutations were generally more common in older patients (aged 60+) vs. younger (38% vs. 15% p < 0.0001), except in CRC (15% vs. 30% q = 0.0001). Black race was associated with non-V600 BRAF alterations (OR: 1.58; p < 0.0001). CONCLUSIONS Class 2/3 BRAFs are more present in Black male patients with co-mutations outside of the MAPK pathway, likely requiring additional oncogenic input for tumorigenesis. Improving access to NGS and trial enrollment will help the development of targeted therapies for non-V600 BRAF mutations.
Collapse
Affiliation(s)
- Suzanne Kazandjian
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H4A 3T2, Canada; (S.K.); (K.M.); (P.K.); (G.B.)
- Segal Cancer Centre, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Emmanuelle Rousselle
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (E.R.); (M.D.)
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Matthew Dankner
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (E.R.); (M.D.)
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - David W. Cescon
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Center, Toronto, ON M5G 2M9, Canada; (D.W.C.); (A.S.)
| | - Anna Spreafico
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Center, Toronto, ON M5G 2M9, Canada; (D.W.C.); (A.S.)
| | - Kim Ma
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H4A 3T2, Canada; (S.K.); (K.M.); (P.K.); (G.B.)
- Segal Cancer Centre, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Petr Kavan
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H4A 3T2, Canada; (S.K.); (K.M.); (P.K.); (G.B.)
- Segal Cancer Centre, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Gerald Batist
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H4A 3T2, Canada; (S.K.); (K.M.); (P.K.); (G.B.)
- Segal Cancer Centre, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - April A. N. Rose
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC H4A 3T2, Canada; (S.K.); (K.M.); (P.K.); (G.B.)
- Segal Cancer Centre, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (E.R.); (M.D.)
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H3G 2M1, Canada
| |
Collapse
|
26
|
Mogavero A, Cantale O, Mollica V, Anpalakhan S, Addeo A, Mountzios G, Friedlaender A, Kanesvaran R, Novello S, Banna GL. First-line immunotherapy in non-small cell lung cancer: how to select and where to go. Expert Rev Respir Med 2023; 17:1191-1206. [PMID: 38294292 DOI: 10.1080/17476348.2024.2302356] [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: 06/11/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024]
Abstract
INTRODUCTION Immunotherapy (IO) has established a new milestone in lung cancer treatment. Several registrational studies have approved immune checkpoint inhibitors (ICIs) in different settings, including the metastatic nonsmall cell lung cancer (NSCLC). As well known, responders are just a certain proportion of patients; therefore, their selection by using predictive factors has stood out as a crucial issue to address in tailoring a patient-centered care. AREAS COVERED In our review we propose a detailed yet handy cross section on ICIs as first-line treatment in metastatic NSCLC, regarding indications, histological, clinical, and blood-based biomarkers, other than their mechanisms of resistance and new immunological actionable targets. We performed a literature search through PubMed entering keywords complying with crucial features of immunotherapy. EXPERT OPINION IO represents the backbone of lung cancer treatment. Trials are currently testing novel immune blockade agents assessing combinatorial approaches with standard ICIs, or antibody drug conjugates (ADC), harboring immunological targets. Perfecting patients' selection is an ongoing challenge and a more and more urgent need in order to best predict responders who will consistently benefit from it.
Collapse
Affiliation(s)
| | | | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Shobana Anpalakhan
- Department of Oncology, Portsmouth Hospitals University NHS Trust, Portsmouth, UK
| | - Alfredo Addeo
- Oncology Department, HUG-Hopitaux Universitaires de Geneve, Geneva, Switzerland
| | - Giannis Mountzios
- Fourth Oncology Department and Clinical Trials Unit, Henry Dunant Hospital Center, Athens, Greece
| | | | - Ravindran Kanesvaran
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Silvia Novello
- Department of Oncology, University of Turin, Turin, Italy
| | - Giuseppe Luigi Banna
- Department of Oncology, Portsmouth Hospitals University NHS Trust, Portsmouth, UK
- Science and Health, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| |
Collapse
|
27
|
Li A, Wang Y, Yu Z, Tan Z, He L, Fu S, Shi M, Du W, Luo L, Li Z, Liu J, Zhou Y, Fang W, Yang Y, Zhang L, Hong S. STK11/LKB1-Deficient Phenotype Rather Than Mutation Diminishes Immunotherapy Efficacy and Represents STING/Type I Interferon/CD8 + T-Cell Dysfunction in NSCLC. J Thorac Oncol 2023; 18:1714-1730. [PMID: 37495171 DOI: 10.1016/j.jtho.2023.07.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/28/2023]
Abstract
INTRODUCTION Conflicting findings have been reported regarding the association between STK11/LKB1 mutations and immune checkpoint inhibitor (ICB) efficacy in NSCLC. It has been reported that tumors could exhibit impaired STK11/LKB1 function even without STK11 mutations. We hypothesized that STK11 phenotype rather than mutation may better stratify ICB outcomes. METHODS Selected functional STK11 events and LKB1 protein data were leveraged to establish a transcriptomics-based classifier of STK11 phenotype (STK11-deficient [-def] or -proficient [-prof]). We analyzed in-house and Genentech/Roche's data of three randomized trials of programmed cell death protein-1 or programmed death-ligand 1 (PD-L1) inhibition in NSCLC (ORIENT-11, n = 171; OAK, n = 699; POPLAR, n = 192) and The Cancer Genome Atlas-NSCLC cohort. RESULTS Tissue STK11 mutation did not affect ICB outcomes. However, the survival benefit of ICB versus chemotherapy were lost or reversed in STK11-def tumors (hazard ratios for death, 95% confidence interval: OAK [0.97, 0.69-1.35]; POPLAR [1.61, 0.88-2.97]; ORIENT-11 [1.07, 0.50-2.29]), while remaining in STK11-prof tumors (hazard ratios for death, 95% confidence interval: OAK [0.81, 0.66-0.99]; POPLAR [0.66, 0.46-0.95]; ORIENT-11 [0.59, 0.37-0.92]). In tumors differentially classified by phenotype and mutation status, STK11-wild-type/def tumors had significantly worse ICB outcomes than STK11-mutated (STK11-MUT)/prof tumors (p < 0.05). The deleterious impact of STK11 deficiency was independent of STK11/KRAS/KEAP1 status or PD-L1 expression. The STING/interferon-I signaling, which was previously shown to be suppressed in STK11-MUT models, was perturbed in patients with STK11-def tumors rather than those with STK11-MUT tumors. Surprisingly, whereas high CD8+ T-cell infiltration was significantly associated with prolonged survival with ICB in STK11-prof tumors (p < 0.05 for 3 trials), it predicted an opposite trend toward worse ICB outcomes in STK11-def tumors across three trials. This suggested an association between STK11 deficiency and CD8+ T-cell dysfunction, which might not be reversed by programmed cell death protein 1 or PD-L1 blockade. CONCLUSIONS STK11 phenotype rather than mutation status can accurately identify patients with ICB-refractory NSCLC and reflect immune suppression. It can help refine stratification algorithms for future clinical research and also provide a reliable resource aiding basic and translational studies in identifying therapeutic targets.
Collapse
Affiliation(s)
- Anlin Li
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yuanyuan Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Zhixin Yu
- State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; Department of VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Zihui Tan
- State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Lina He
- State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Sha Fu
- Department of Cellular and Molecular Diagnostics Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Mengting Shi
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wei Du
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Linfeng Luo
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Zhichao Li
- State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jiaqing Liu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yixin Zhou
- State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; Department of VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Wenfeng Fang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yunpeng Yang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Li Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Shaodong Hong
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China; State Key Laboratory of Oncology in South People's Republic of China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China.
| |
Collapse
|
28
|
Jun S, Park S, Sun JM, Lee SH, Ahn JS, Ahn MJ, Cho J, Jung HA. Real-World Outcomes of Immunotherapy in Second- or Later-Line Non-Small Cell Lung Cancer with Actionable Genetic Alterations. Cancers (Basel) 2023; 15:5450. [PMID: 38001710 PMCID: PMC10670305 DOI: 10.3390/cancers15225450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
INTRODUCTION While the efficacy of immune checkpoint inhibitors (ICIs) in treating non-small cell lung cancer (NSCLC) patients with actionable genetic alterations (AGAs) is modest, certain patients demonstrate improved survival. Thus, this study aimed to evaluate the benefits of ICIs in NSCLC patients with diverse AGAs and verify the predictive biomarkers of ICI efficacy. METHODS From January 2018 to July 2022, this study compared the progression-free survival (PFS) of NSCLC patients with different AGAs treated with ICI monotherapy as second- or later-line therapy at Samsung Medical Center. To ascertain the predictors of ICIs efficacy, we adjusted ICIs' effects on PFS in terms of clinical and molecular biomarkers. RESULTS EGFR (46.0%) was the most prevalent mutation in 324 patients. In multivariate analysis, PD-L1 positivity (tumor proportion score (TPS) ≥ 1%) (HR = 0.41) and the use of steroids for immune-related adverse events (HR = 0.46) were positive factors for ICI therapy in the AGAs group. Co-existing mutation of STK11 with KRAS mutation (HR = 4.53) and TP53 with MET mutation (HR = 9.78) was negatively associated with survival. CONCLUSIONS The efficacy of ICI treatment varied across AGA subtypes, but patients with KRAS, MET, and BRAF mutations demonstrated relatively long-duration benefits of ICI therapy. PD-L1 was a significant positive predictive biomarker in all AGA groups.
Collapse
Affiliation(s)
- Soojin Jun
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul 06355, Republic of Korea; (S.J.); (J.C.)
| | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; (S.P.); (J.-M.S.); (S.-H.L.); (J.S.A.); (M.-J.A.)
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; (S.P.); (J.-M.S.); (S.-H.L.); (J.S.A.); (M.-J.A.)
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; (S.P.); (J.-M.S.); (S.-H.L.); (J.S.A.); (M.-J.A.)
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; (S.P.); (J.-M.S.); (S.-H.L.); (J.S.A.); (M.-J.A.)
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; (S.P.); (J.-M.S.); (S.-H.L.); (J.S.A.); (M.-J.A.)
| | - Juhee Cho
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul 06355, Republic of Korea; (S.J.); (J.C.)
| | - Hyun Ae Jung
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea; (S.P.); (J.-M.S.); (S.-H.L.); (J.S.A.); (M.-J.A.)
| |
Collapse
|
29
|
Marchal M, Leroy V, Behal H, Dansin E, Paris N, Bordier S, Humez S, Escande F, Gauvain C, Cortot AB. Histo-Molecular Factors of Response to Combined Chemotherapy and Immunotherapy in Non-Small Cell Lung Cancers. Target Oncol 2023; 18:927-939. [PMID: 37921939 PMCID: PMC10663251 DOI: 10.1007/s11523-023-01009-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 11/05/2023]
Abstract
BACKGROUND Chemo-immunotherapy (CIT) is the standard of care for advanced non-small cell lung cancer (NSCLC), but the impact of routinely available histo-molecular biomarkers on its efficacy has not yet been fully assessed. OBJECTIVE The purpose of this multicenter study was to evaluate the clinical activity of CIT according to oncogenic drivers, STK11 and TP53 mutations, and MET overexpression. PATIENTS AND METHODS Patients receiving CIT for advanced NSCLC with available comprehensive molecular profile were included. The primary endpoint was progression-free survival (PFS), adjusted on main confounding factors, and secondary endpoints were overall survival (OS) and objective response rate. RESULTS Among the 195 patients included between September 2018 and October 2021, 88 (41%) had a KRAS mutation, 16 (8.2%) an EGFR mutation or an ALK, ROS1, or RET rearrangement, 11 (5.6%) a BRAF mutation, 6 (3.1%) a MET exon 14 mutation or MET amplification, and 5 (2.6%) a HER2 mutation. Seventy-seven patients (39.5%) had none of these alterations. The median PFS was 6.4 months (95% CI 5.3-7.3). Per subgroup, the median PFS was 7.1 months (5.4-8.9) for KRAS, 5.5 months (2.5-15.3) for EGFR/ALK/ROS1/RET, 12.9 months (2.6-not reached [NR]) for BRAF, 1.5 months (0.6-NR) for MET, 3.9 months (2.6-NR) for HER2, and 5.6 months (4.7-7.8) for patients without any oncogenic alteration. No difference in PFS was observed between the KRAS, BRAF, EGFR/ALK/ROS1/RET, and no-driver subgroups. STK11 mutations were associated with poor PFS (HR 1.59 [95% CI 1.01-2.51]) whereas TP53 mutations had no impact. MET overexpression was associated with longer PFS (HR 0.59 [95% CI 0.35-0.99]). CONCLUSION This study suggests that the efficacy of combining pembrolizumab with pemetrexed and platinum-based chemotherapy differs according to the histo-molecular biomarkers, which may help to identify patients liable to benefit from CIT.
Collapse
Affiliation(s)
- Marine Marchal
- Department of Thoracic Oncology, Univ. Lille, CHU Lille, Boulevard du Professeur Leclercq, 59000, Lille, France.
| | - Vincent Leroy
- Department of Pneumology, Clinique Tessier, Valenciennes, France
| | - Hélène Behal
- Biostatistics Department, CHU Lille, 59000, Lille, France
| | - Eric Dansin
- Department of Medical Oncology, Centre Oscar Lambret, Lille, France
| | - Nicolas Paris
- Department of Pneumology, Dunkerque Hospital, Dunkerque, France
| | - Soraya Bordier
- Department of Pneumology, Roubaix Hospital, Roubaix, France
| | - Sarah Humez
- Department of Pathology, CHU Lille, Univ. Lille, CHU de Lille, 59000, Lille, France
- CNRS, Inserm, Institut Pasteur de Lille, UMR9020-UMR1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Univ. Lille, CHU Lille, 59000, Lille, France
| | - Fabienne Escande
- Department of Biochemistry and Molecular Biology, CHU Lille, Univ. Lille, CHU de Lille, 59000, Lille, France
| | - Clément Gauvain
- Department of Thoracic Oncology, Univ. Lille, CHU Lille, Boulevard du Professeur Leclercq, 59000, Lille, France
| | - Alexis B Cortot
- Department of Thoracic Oncology, Univ. Lille, CHU Lille, Boulevard du Professeur Leclercq, 59000, Lille, France
- CNRS, Inserm, Institut Pasteur de Lille, UMR9020-UMR1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Univ. Lille, CHU Lille, 59000, Lille, France
| |
Collapse
|
30
|
Sumii M, Namba M, Tokumo K, Yamauchi M, Okamoto W, Hattori N, Sugiyama K. Concurrent Mutations in STK11 and KEAP1 Cause Treatment Resistance in KRAS Wild-type Non-small-cell Lung Cancer. Intern Med 2023; 62:3001-3004. [PMID: 36858519 PMCID: PMC10641186 DOI: 10.2169/internalmedicine.1110-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/20/2023] [Indexed: 03/03/2023] Open
Abstract
We herein report a patient with KRAS wild-type non-small-cell lung cancer (NSCLC) with concurrent STK11 and KEAP1 mutations. A 53-year-old man visited a local doctor with a complaint of left shoulder swelling and pain. He was diagnosed with NSCLC cT4N0M1c stage IVB. A comprehensive genome profile test revealed mutations in STK11 and KEAP1 but no KRAS mutations. The patient was refractory to radiotherapy, immunotherapy, and chemotherapy. Thus, STK11 and KEAP1 mutations can be considered resistance mutations that confer resistance to various anticancer therapies in KRAS wild-type NSCLC.
Collapse
Affiliation(s)
- Masahiko Sumii
- Department of Respiratory Medicine, Hiroshima University Hospital, Japan
| | - Masashi Namba
- Department of Clinical Oncology, Hiroshima University Hospital, Japan
| | - Kentaro Tokumo
- Department of Clinical Oncology, Hiroshima University Hospital, Japan
| | - Masami Yamauchi
- Department of Clinical Oncology, Hiroshima University Hospital, Japan
| | - Wataru Okamoto
- Department of Cancer Treatment Center, Hiroshima University Hospital, Japan
| | - Noboru Hattori
- Department of Respiratory Medicine, Hiroshima University Hospital, Japan
| | - Kazuhiko Sugiyama
- Department of Clinical Oncology, Hiroshima University Hospital, Japan
- Department of Cancer Treatment Center, Hiroshima University Hospital, Japan
| |
Collapse
|
31
|
Boeschen M, Kuhn CK, Wirtz H, Seyfarth HJ, Frille A, Lordick F, Hacker UT, Obeck U, Stiller M, Bläker H, von Laffert M. Comparative bioinformatic analysis of KRAS, STK11 and KEAP1 (co-)mutations in non-small cell lung cancer with a special focus on KRAS G12C. Lung Cancer 2023; 184:107361. [PMID: 37699269 DOI: 10.1016/j.lungcan.2023.107361] [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: 07/25/2023] [Accepted: 09/04/2023] [Indexed: 09/14/2023]
Abstract
OBJECTIVES Mutations in STK11 (STK11MUT) and KEAP1 (KEAP1MUT) occur frequently in non-small cell lung cancer (NSCLC) and are often co-mutated with KRAS. Several studies linked the co-occurrence of KRASMUT + STK11MUT, as well as KRASMUT + KEAP1MUT to reduced response to immune checkpoint inhibitors (ICI) and even a negative impact on survival. Data focusing STK11 + KEAP1 co-mutations or the triple mutation (KRAS + STK11 + KEAP1) are scarce. The recent availability of KRAS-G12C inhibitors increases the clinical relevance of those co-mutations in KRAS-mutated NSCLC. MATERIALS AND METHODS We present a comprehensive bioinformatic analysis encompassing six datasets retrieved from cBioPortal. RESULTS Independent of the treatment, triple mutations and STK11MUT + KEAP1MUT were significantly associated with a reduced overall survival (OS). Across treatments, OS of patients with a KRAS G12C triple mutation was significantly reduced compared to patients with KRAS G12C-only. Under ICI-therapy, there was no significant difference in OS between patients harboring the KRAS G12C-only and patients with the KRAS G12C triple mutation, but a significant difference between patients harboring KRAS non-G12C and KRAS non-G12C triple mutations. Triple mutated primary tumors showed a significantly increased frequency of distant metastases to bone and adrenal glands compared to KRAS-only mutated tumors. Additionally, our drug response analysis in cancer cell lines harboring the triple mutations revealed the WNT pathway inhibitor XAV-939 as a potential future drug candidate for this mutational situation. CONCLUSION The triple mutation status may serve as a negative prognostic and predictive factor across treatments compared to KRASMUT-only. KRAS G12C generally seems to be a negative predictive marker for ICI-therapy.
Collapse
Affiliation(s)
- Myriam Boeschen
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany.
| | - Christina Katharina Kuhn
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| | - Hubert Wirtz
- Department of Respiratory Medicine, Leipzig University Medical Center, Liebigstrasse 20, 04103 Leipzig
| | - Hans-Jürgen Seyfarth
- Department of Respiratory Medicine, Leipzig University Medical Center, Liebigstrasse 20, 04103 Leipzig
| | - Armin Frille
- Department of Respiratory Medicine, Leipzig University Medical Center, Liebigstrasse 20, 04103 Leipzig
| | - Florian Lordick
- Department of Medicine II, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Leipzig, Germany
| | - Ulrich T Hacker
- Department of Medicine II, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Leipzig, Germany
| | - Ulrike Obeck
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany
| | - Mathias Stiller
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany
| | - Hendrik Bläker
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany
| | - Maximilian von Laffert
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany.
| |
Collapse
|
32
|
Wankhede D, Bontoux C, Grover S, Hofman P. Prognostic Role of KRAS G12C Mutation in Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis. Diagnostics (Basel) 2023; 13:3043. [PMID: 37835787 PMCID: PMC10572143 DOI: 10.3390/diagnostics13193043] [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: 08/17/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/15/2023] Open
Abstract
KRAS G12C mutation (mKRAS G12C) is the most frequent KRAS point mutation in non-small cell lung cancer (NSCLC) and has been proven to be a predictive biomarker for direct KRAS G12C inhibitors in advanced solid cancers. We sought to determine the prognostic significance of mKRAS G12C in patients with NSCLC using the meta-analytic approach. A protocol is registered at the International Prospective Register for systematic reviews (CRD42022345868). PubMed, EMBASE, The Cochrane Library, and Clinicaltrials.gov.in were searched for prospective or retrospective studies reporting survival data for tumors with mKRAS G12C compared with either other KRAS mutations or wild-type KRAS (KRAS-WT). The hazard ratios (HRs) for overall survival (OS) or Disease-free survival (DFS) of tumors were pooled according to fixed or random-effects models. Sixteen studies enrolling 10,153 participants were included in the final analysis. mKRAS G12C tumors had poor OS [HR, 1.42; 95% CI, 1.10-1.84, p = 0.007] but similar DFS [HR 2.36, 95% CI 0.64-8.16] compared to KRAS-WT tumors. Compared to other KRAS mutations, mKRAS G12C tumors had poor DFS [HR, 1.49; 95% CI, 1.07-2.09, p < 0.0001] but similar OS [HR, 1.03; 95% CI, 0.84-1.26]. Compared to other KRAS mutations, high PD-L1 expression (>50%) [OR 1.37 95% CI 1.11-1.70, p = 0.004] was associated with mKRAS G12C tumors. mKRAS G12C is a promising prognostic factor for patients with NSCLC, negatively impacting survival. Prevailing significant heterogeneity and selection bias might reduce the validity of these findings. Concomitant high PD-L1 expression in these tumors opens doors for exciting therapeutic potential.
Collapse
Affiliation(s)
- Durgesh Wankhede
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Christophe Bontoux
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Centre Hospitalier, Université Côte d’Azur, 06002 Nice, France;
| | - Sandeep Grover
- Centre for Genetic Epidemiology, Institute for Clinical Epidemiology and Applied Biometry, University of Tübingen, 72076 Tübingen, Germany;
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Centre Hospitalier, Université Côte d’Azur, 06002 Nice, France;
- Institute for Research on Cancer and Ageing, Nice (IRCAN), INSERM U1081 and UMR CNRS 7284, Team 4, 06107 Nice, France;
- Hospital-Integrated Biobank BB-0033-00025, Pasteur Hospital, 06000 Nice, France
- University Hospital Federation OncoAge, CHU de Nice, University Côte d’Azur, 06000 Nice, France
| |
Collapse
|
33
|
Roque K, Ruiz R, Mas L, Pozza DH, Vancini M, Silva Júnior JA, de Mello RA. Update in Immunotherapy for Advanced Non-Small Cell Lung Cancer: Optimizing Treatment Sequencing and Identifying the Best Choices. Cancers (Basel) 2023; 15:4547. [PMID: 37760516 PMCID: PMC10526179 DOI: 10.3390/cancers15184547] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/09/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
The introduction of immunotherapy has brought about a paradigm shift in the management of advanced non-small cell lung cancer (NSCLC). It has not only significantly improved the prognosis of patients but has also become a cornerstone of treatment, particularly in those without oncogenic driver mutations. Immune checkpoint inhibitors (ICIs) play a crucial role in the treatment of lung cancer and can be classified into two main groups: Anti-cytotoxic T lymphocyte antigen-4 (Anti-CTLA-4) and anti-T-cell receptor programmed cell death-1 or its ligand (Anti-PD-1 and Anti-PD-L1). Certainly, the landscape of approved first line immunotherapeutic approaches has expanded to encompass monotherapy, immunotherapy-exclusive protocols, and combinations with chemotherapy. The complexity of decision-making in this realm arises due to the absence of direct prospective comparisons. However, a thorough analysis of the long-term efficacy and safety data derived from pivotal clinical trials can offer valuable insights into optimizing treatment for different patient subsets. Moreover, ongoing research is investigating emerging biomarkers and innovative therapeutic strategies that could potentially refine the current treatment approach even further. In this comprehensive review, our aim is to highlight the latest advances in immunotherapy for advanced NSCLC, including the mechanisms of action, efficacy, safety profiles, and clinical significance of ICI.
Collapse
Affiliation(s)
- Katia Roque
- Discipline of Medical Oncology, Post-Graduation Programme in Medicine, Faculty of Medicine, Nine of July University (UNINOVE), São Paulo 04101-000, Brazil (J.A.S.J.)
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Angamos Este Av., 2520, Lima 15023, Peru; (R.R.); (L.M.)
- Faculty of Medicine, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
- Faculty of Medicine, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru
| | - Rossana Ruiz
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Angamos Este Av., 2520, Lima 15023, Peru; (R.R.); (L.M.)
- Escuela Profesional de Medicina Humana-Filial Ica, Universidad Privada San Juan Bautista, Ica 15067, Peru
| | - Luis Mas
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Angamos Este Av., 2520, Lima 15023, Peru; (R.R.); (L.M.)
- Faculty of Medicine, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
- Department of Medical Oncology, Oncosalud-AUNA, Av. Guardia Civil 571-San Borja, Lima 15036, Peru
| | - Daniel Humberto Pozza
- Experimental Biology Unit, Department of Biomedicine, Faculty of Medicine of Porto, University of Porto, 4200-319 Porto, Portugal
- i3S—Institute for Research and Innovation in Health and IBMC, University of Porto, 4200-319 Porto, Portugal
| | - Marina Vancini
- Discipline of Medical Oncology, Post-Graduation Programme in Medicine, Faculty of Medicine, Nine of July University (UNINOVE), São Paulo 04101-000, Brazil (J.A.S.J.)
| | - José Antônio Silva Júnior
- Discipline of Medical Oncology, Post-Graduation Programme in Medicine, Faculty of Medicine, Nine of July University (UNINOVE), São Paulo 04101-000, Brazil (J.A.S.J.)
| | - Ramon Andrade de Mello
- Discipline of Medical Oncology, Post-Graduation Programme in Medicine, Faculty of Medicine, Nine of July University (UNINOVE), São Paulo 04101-000, Brazil (J.A.S.J.)
- Oxford Cancer Centre, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
- Department of Oncology, University of Oxford, Oxford OX1 2JD, UK
| |
Collapse
|
34
|
Di Federico A, Ricciotti I, Favorito V, Michelina SV, Scaparone P, Metro G, De Giglio A, Pecci F, Lamberti G, Ambrogio C, Ricciuti B. Resistance to KRAS G12C Inhibition in Non-small Cell Lung Cancer. Curr Oncol Rep 2023; 25:1017-1029. [PMID: 37378881 DOI: 10.1007/s11912-023-01436-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2023] [Indexed: 06/29/2023]
Abstract
PURPOSE OF REVIEW Although the recent development of direct KRASG12C inhibitors (G12Ci) has improved outcomes in KRAS mutant cancers, responses occur only in a fraction of patients, and among responders acquired resistance invariably develops over time. Therefore, the characterization of the determinants of acquired resistance is crucial to inform treatment strategies and to identify novel therapeutic vulnerabilities that can be exploited for drug development. RECENT FINDINGS Mechanisms of acquired resistance to G12Ci are heterogenous including both on-target and off-target resistance. On-target acquired resistance includes secondary codon 12 KRAS mutations, but also acquired codon 13 and codon 61 alterations, and mutations at drug binding sites. Off-target acquired resistance can derive from activating mutations in KRAS downstream pathway (e.g., MEK1), acquired oncogenic fusions (EML4-ALK, CCDC176-RET), gene level copy gain (e.g., MET amplification), or oncogenic alterations in other pro-proliferative and antiapoptotic pathways (e.g., FGFR3, PTEN, NRAS). In a fraction of patients, histologic transformation can also contribute to the development of acquire resistance. We provided a comprehensive overview of the mechanisms that limit the efficacy of this G12i and reviewed potential strategies to overcome and possibly delay the development of resistance in patients receiving KRAS directed targeted therapies.
Collapse
Affiliation(s)
- Alessandro Di Federico
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA.
- Department of Medical and Surgical Sciences, University of Bologna, Via Albertoni, 15, 40138, Bologna, Italy.
| | - Ilaria Ricciotti
- Department of Medical and Surgical Sciences, University of Bologna, Via Albertoni, 15, 40138, Bologna, Italy
| | - Valentina Favorito
- Department of Medical and Surgical Sciences, University of Bologna, Via Albertoni, 15, 40138, Bologna, Italy
| | - Sandra Vietti Michelina
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology, Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Pietro Scaparone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology, Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Giulio Metro
- Medical Oncology, Santa Maria Della Misericordia Hospital, Azienda Ospedaliera di Perugia, Piazzale Giorgio Menghini, 1, 06129, Perugia, Italy
| | - Andrea De Giglio
- Department of Medical and Surgical Sciences, University of Bologna, Via Albertoni, 15, 40138, Bologna, Italy
| | - Federica Pecci
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Giuseppe Lamberti
- Department of Medical and Surgical Sciences, University of Bologna, Via Albertoni, 15, 40138, Bologna, Italy
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology, Center, University of Torino, Via Nizza 52, 10126, Torino, Italy
| | - Biagio Ricciuti
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA.
| |
Collapse
|
35
|
Cai R, Zhu H, Liu Y, Sha H, Peng W, Yin R, Zhou G, Fang Y. To be, or not to be: the dilemma of immunotherapy for non-small cell lung cancer harboring various driver mutations. J Cancer Res Clin Oncol 2023; 149:10027-10040. [PMID: 37261523 PMCID: PMC10423141 DOI: 10.1007/s00432-023-04919-4] [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: 03/23/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
INTRODUCTION Lung cancer is one of primary cancer type with high incidence and mortality, non-small cell lung cancer (NSCLC) is the most common type of lung cncer. For advanced lung cancer, traditional chemotherapy and targeted therapy become difficult to solve the dilemma of further progress. In recent years, with the clinical application of immunotherapy, the therapeutic strategy of lung cancer has changed dramatically. At present, immunotherapy has shown conspicuous efficacy in NSCLC patients with high expression of programmed death-ligand 1 (PD-L1) and high tumor mutational burden (TMB). The discovery of driver mutations brings delightful hope for targeted cancer therapy. However, it remains controversial whether immunotherapy can be used in NSCLC patients with these specific driver mutations. METHOD This article summarized the latest research progresses of immunotherapy in advanced NSCLC. We paid close attention to the relevance of various driver mutations and immunotherapy in NSCLC patients, and summarized the predictive effects of several driver mutations and immunotherapy. RESULTS The mutations of KRAS, KRAS+TP53, EPHA (especially EPHA5), ZFHX3, ZFHX3+TP53, NOTCH, BRAF and LRP1B+FAT3 have potential to be used as biomarkers to predict the positive effectiveness of immunotherapy. ZFHX3, ZFHX3+TP53, STKII/LKB1+KEAP1+SMARCA4+PBRM1 mutations in LUAD patients get more positive effect in immunotherapy. While the mutations of EGFR, KEAP1, STKII/LKB1+KRAS, EML4-ALK, MET exon 14 skipping mutation, PBRM1, STKII/LKB1+KEAP1+SMARCA4+PBRM1, ERBB2, PIK3CA and RET often indicate poor benefit from immunotherapy. CONCLUSION Many gene mutations have been shown to be associated with immunotherapy efficacy. Gene mutations should be combined with PD-L1, TMB, etc. to predict the effect of immunotherapy.
Collapse
Affiliation(s)
- Ruoxue Cai
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Baiziting 42, Nanjing, 210009, People's Republic of China
| | - Hongyu Zhu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, 210009, People's Republic of China
| | - Ying Liu
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Baiziting 42, Nanjing, 210009, People's Republic of China
| | - Huanhuan Sha
- Department of Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, People's Republic of China
| | - Weiwei Peng
- Department of Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, People's Republic of China
| | - Rong Yin
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Jiangsu Cancer Hospital & Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Institute of Cancer Research, Nanjing, 210009, People's Republic of China
| | - Guoren Zhou
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Baiziting 42, Nanjing, 210009, People's Republic of China.
| | - Ying Fang
- Department of Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, People's Republic of China
| |
Collapse
|
36
|
TeSlaa T, Ralser M, Fan J, Rabinowitz JD. The pentose phosphate pathway in health and disease. Nat Metab 2023; 5:1275-1289. [PMID: 37612403 PMCID: PMC11251397 DOI: 10.1038/s42255-023-00863-2] [Citation(s) in RCA: 173] [Impact Index Per Article: 86.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 07/12/2023] [Indexed: 08/25/2023]
Abstract
The pentose phosphate pathway (PPP) is a glucose-oxidizing pathway that runs in parallel to upper glycolysis to produce ribose 5-phosphate and nicotinamide adenine dinucleotide phosphate (NADPH). Ribose 5-phosphate is used for nucleotide synthesis, while NADPH is involved in redox homoeostasis as well as in promoting biosynthetic processes, such as the synthesis of tetrahydrofolate, deoxyribonucleotides, proline, fatty acids and cholesterol. Through NADPH, the PPP plays a critical role in suppressing oxidative stress, including in certain cancers, in which PPP inhibition may be therapeutically useful. Conversely, PPP-derived NADPH also supports purposeful cellular generation of reactive oxygen species (ROS) and reactive nitrogen species (RNS) for signalling and pathogen killing. Genetic deficiencies in the PPP occur relatively commonly in the committed pathway enzyme glucose-6-phosphate dehydrogenase (G6PD). G6PD deficiency typically manifests as haemolytic anaemia due to red cell oxidative damage but, in severe cases, also results in infections due to lack of leucocyte oxidative burst, highlighting the dual redox roles of the pathway in free radical production and detoxification. This Review discusses the PPP in mammals, covering its roles in biochemistry, physiology and disease.
Collapse
Affiliation(s)
- Tara TeSlaa
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Markus Ralser
- Department of Biochemistry, Charité Universitätsmedizin, Berlin, Germany
- The Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Jing Fan
- Morgride Institute for Research, Madison, WI, USA
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Joshua D Rabinowitz
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
- Department of Chemistry, Princeton University, Princeton, NJ, USA.
- Ludwig Institute for Cancer Research, Princeton Branch, Princeton, NJ, USA.
| |
Collapse
|
37
|
Cheema PK, Banerji SO, Blais N, Chu QSC, Juergens RA, Leighl NB, Sacher A, Sheffield BS, Snow S, Vincent M, Wheatley-Price PF, Yip S, Melosky BL. Canadian Consensus Recommendations on the Management of KRAS G12C-Mutated NSCLC. Curr Oncol 2023; 30:6473-6496. [PMID: 37504336 PMCID: PMC10377814 DOI: 10.3390/curroncol30070476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 07/29/2023] Open
Abstract
Activating mutations in Kirsten rat sarcoma viral oncogene homologue (KRAS), in particular, a point mutation leading to a glycine-to-cysteine substitution at codon 12 (G12C), are among the most frequent genomic alterations in non-small cell lung cancer (NSCLC). Several agents targeting KRAS G12C have recently entered clinical development. Sotorasib, a first-in-class specific small molecule that irreversibly inhibits KRAS G12C, has since obtained Health Canada approval. The emergence of novel KRAS-targeted therapies warrants the development of evidence-based consensus recommendations to help clinicians better understand and contextualize the available data. A Canadian expert panel was convened to define the key clinical questions, review recent evidence, and discuss and agree on recommendations for the treatment of advanced KRAS G12C-mutated NSCLC. The panel agreed that testing for KRAS G12C should be performed as part of a comprehensive panel that includes current standard-of-care biomarkers. Sotorasib, the only approved KRAS G12C inhibitor in Canada, is recommended for patients with advanced KRAS G12C-mutated NSCLC who progressed on guideline-recommended first-line standard of care for advanced NSCLC without driver alterations (immune-checkpoint inhibitor(s) [ICIs] +/- chemotherapy). Sotorasib could also be offered as second-line therapy to patients who progressed on ICI monotherapy that are not candidates for a platinum doublet and those that received first-line chemotherapy with a contraindication to ICIs. Preliminary data indicate the activity of KRAS G12C inhibitors in brain metastases; however, the evidence is insufficient to make specific recommendations. Regular liver function monitoring is recommended when patients are prescribed KRAS G12C inhibitors due to risk of hepatotoxicity.
Collapse
Affiliation(s)
- Parneet K. Cheema
- Division of Medical Oncology, William Osler Health System, University of Toronto, Brampton, ON L6R 3J7, Canada
- Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Shantanu O. Banerji
- CancerCare Manitoba Research Institute, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0V9, Canada;
| | - Normand Blais
- Department of Medicine, Centre Hospitalier de l’Université de Montréal, University of Montreal, Montreal, QC H2X 3E4, Canada;
| | - Quincy S.-C. Chu
- Division of Medical Oncology, Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
| | - Rosalyn A. Juergens
- Department of Medical Oncology, Juravinski Cancer Centre, McMaster University, Hamilton, ON L8V 5C2, Canada;
| | - Natasha B. Leighl
- Department of Medicine, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5S 1A8, Canada; (N.B.L.); (A.S.)
| | - Adrian Sacher
- Department of Medicine, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON M5S 1A8, Canada; (N.B.L.); (A.S.)
| | - Brandon S. Sheffield
- Department of Laboratory Medicine, William Osler Health System, Brampton, ON L6R 3J7, Canada
| | - Stephanie Snow
- Division of Medical Oncology, Department of Medicine, QEII Health Sciences Centre, Dalhousie University, Halifax, NS B3H 2Y9, Canada;
| | - Mark Vincent
- Department of Medical Oncology, London Regional Cancer Program, London, ON N6A 5W9, Canada;
| | - Paul F. Wheatley-Price
- Department of Medicine, The Ottawa Hospital Research Institute, The Ottawa Hospital, University of Ottawa, Ottawa, ON K1H 8L6, Canada;
| | - Stephen Yip
- BC Cancer, Vancouver, University of British Columbia, Vancouver, BC V6T 1Z4, Canada;
| | - Barbara L. Melosky
- Department of Medical Oncology, BC Cancer-Vancouver Centre, Vancouver, BC V5Z 4E6, Canada;
| |
Collapse
|
38
|
Zhou F, Zhou C. Notch Mutations as a Novel Biomarker for Immunotherapy. J Thorac Oncol 2023; 18:e54-e56. [PMID: 37087126 DOI: 10.1016/j.jtho.2023.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 04/24/2023]
Affiliation(s)
- Fei Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
39
|
Mohanty SK, Mishra SK, Amin MB, Agaimy A, Fuchs F. Role of Surgical Pathologist for the Detection of Immuno-oncologic Predictive Factors in Non-small Cell Lung Cancers. Adv Anat Pathol 2023; 30:174-194. [PMID: 37037418 DOI: 10.1097/pap.0000000000000395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
Until very recently, surgery, chemotherapy, and radiation therapy have been the mainstay of treatment in non-small cell carcinomas (NSCLCs). However, recent advances in molecular immunology have unveiled some of the complexity of the mechanisms regulating cellular immune responses and led to the successful targeting of immune checkpoints in attempts to enhance antitumor T-cell responses. Immune checkpoint molecules such as cytotoxic T-lymphocyte associated protein-4, programmed cell death protein-1, and programmed death ligand (PD-L) 1 have been shown to play central roles in evading cancer immunity. Thus, these molecules have been targeted by inhibitors for the management of cancers forming the basis of immunotherapy. Advanced NSCLC has been the paradigm for the benefits of immunotherapy in any cancer. Treatment decisions are made based on the expression of PD-L1 on the tumor cells and the presence or absence of driver mutations. Patients with high PD-L1 expression (≥50%) and no driver mutations are treated with single-agent immunotherapy whereas, for all other patients with a lower level of PD-L1 expression, a combination of chemotherapy and immunotherapy is preferred. Thus, PD-L1 blockers are the only immunotherapeutic agents approved in advanced NSCLC without any oncogenic driver mutations. PD-L1 immunohistochemistry, however, may not be the best biomarker in view of its dynamic nature in time and space, and the benefits may be seen regardless of PD -L1 expression. Each immunotherapy molecule is prescribed based on the levels of PD-L1 expression as assessed by a Food and Drug Administration-approved companion diagnostic assay. Other biomarkers that have been studied include tumor mutational burden, the T-effector signature, tumor-infiltrating lymphocytes, radiomic assays, inflammation index, presence or absence of immune-related adverse events and specific driver mutations, and gut as well as local microbiome. At the current time, none of these biomarkers are routinely used in the clinical decision-making process for immunotherapy in NSCLC. However, in individual cases, they can be useful adjuncts to conventional therapy. This review describes our current understanding of the role of biomarkers as predictors of response to immune checkpoint molecules. To begin with a brief on cancer immunology in general and in NSCLC, in particular, is discussed. In the end, recent advancements in laboratory techniques for refining biomarker assays are described.
Collapse
Affiliation(s)
- Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, Advanced Medical Research Institute, Bhubaneswar, India and CORE Diagnostics, Gurgaon, HR
| | - Sourav K Mishra
- Department of Medical Oncology, All India Institute of Medical Sciences, DL, India
| | - Mahul B Amin
- Departments of Pathology and Laboratory Medicine and Urology, University of Tennessee Health Science Center, Memphis, TN
| | - Abbas Agaimy
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Florian Fuchs
- Department of Internal Medicine-1, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Erlangen University Hospital and Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| |
Collapse
|
40
|
Hendriks LE, Kerr KM, Menis J, Mok TS, Nestle U, Passaro A, Peters S, Planchard D, Smit EF, Solomon BJ, Veronesi G, Reck M. Non-oncogene-addicted metastatic non-small-cell lung cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2023; 34:358-376. [PMID: 36669645 DOI: 10.1016/j.annonc.2022.12.013] [Citation(s) in RCA: 271] [Impact Index Per Article: 135.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 01/18/2023] Open
Affiliation(s)
- L E Hendriks
- Department of Pulmonology, GROW School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - K M Kerr
- Aberdeen Royal Infirmary, Aberdeen University Medical School, Aberdeen, UK
| | - J Menis
- Medical Oncology Department, University and Hospital Trust of Verona, Verona, Italy
| | - T S Mok
- Department of Clinical Oncology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - U Nestle
- Department of Radiation Oncology, University Hospital Freiburg, Freiburg; Department of Radiation Oncology, Kliniken Maria Hilf, Moenchengladbach, Germany
| | - A Passaro
- Division of Thoracic Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - S Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne University, Lausanne, Switzerland
| | - D Planchard
- Department of Medical Oncology, Thoracic Group, Gustave-Roussy, Villejuif, France
| | - E F Smit
- Thoracic Oncology Service, Netherlands Cancer Institute, Amsterdam; Department of Pulmonary Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - B J Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - G Veronesi
- Faculty of Medicine and Surgery-Vita-Salute San Raffaele University, Milan; Division of Thoracic Surgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - M Reck
- Department of Thoracic Oncology, Airway Research Center North, German Center for Lung Research, Lung Clinic, Grosshansdorf, Germany
| |
Collapse
|
41
|
Watterson A, Coelho MA. Cancer immune evasion through KRAS and PD-L1 and potential therapeutic interventions. Cell Commun Signal 2023; 21:45. [PMID: 36864508 PMCID: PMC9979509 DOI: 10.1186/s12964-023-01063-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/31/2023] [Indexed: 03/04/2023] Open
Abstract
Oncogenic driver mutations have implications that extend beyond cancer cells themselves. Aberrant tumour cell signalling has various effects on the tumour microenvironment and anti-tumour immunity, with important consequences for therapy response and resistance. We provide an overview of how mutant RAS, one of the most prevalent oncogenic drivers in cancer, can instigate immune evasion programs at the tumour cell level and through remodelling interactions with the innate and adaptive immune cell compartments. Finally, we describe how immune evasion networks focused on RAS, and the immune checkpoint molecule PD-L1 can be disrupted through therapeutic intervention, and discuss potential strategies for combinatorial treatment. Video abstract.
Collapse
Affiliation(s)
- Alex Watterson
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK.,Open Targets, Cambridge, UK
| | - Matthew A Coelho
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK. .,Open Targets, Cambridge, UK.
| |
Collapse
|
42
|
Russano M, La Cava G, Cortellini A, Citarella F, Galletti A, Di Fazio GR, Santo V, Brunetti L, Vendittelli A, Fioroni I, Pantano F, Tonini G, Vincenzi B. Immunotherapy for Metastatic Non-Small Cell Lung Cancer: Therapeutic Advances and Biomarkers. Curr Oncol 2023; 30:2366-2387. [PMID: 36826142 PMCID: PMC9955173 DOI: 10.3390/curroncol30020181] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Immunotherapy has revolutionized the treatment paradigm of non-small cell lung cancer and improved patients' prognosis. Immune checkpoint inhibitors have quickly become standard frontline treatment for metastatic non-oncogene addicted disease, either as a single agent or in combination strategies. However, only a few patients have long-term benefits, and most of them do not respond or develop progressive disease during treatment. Thus, the identification of reliable predictive and prognostic biomarkers remains crucial for patient selection and guiding therapeutic choices. In this review, we provide an overview of the current strategies, highlighting the main clinical challenges and novel potential biomarkers.
Collapse
Affiliation(s)
- Marco Russano
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Giulia La Cava
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Alessio Cortellini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Fabrizio Citarella
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Alessandro Galletti
- Division of Medical Oncology, San Camillo Forlanini Hospital, 00152 Roma, Italy
| | - Giuseppina Rita Di Fazio
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Valentina Santo
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Leonardo Brunetti
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Alessia Vendittelli
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Iacopo Fioroni
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Francesco Pantano
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Giuseppe Tonini
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| | - Bruno Vincenzi
- Department of Medical Oncology, Campus Bio-Medico University of Rome, Via Álvaro del Portillo, 21, 00128 Rome, Italy
| |
Collapse
|
43
|
Zhou K, Li S, Zhao Y, Cheng K. Mechanisms of drug resistance to immune checkpoint inhibitors in non-small cell lung cancer. Front Immunol 2023; 14:1127071. [PMID: 36845142 PMCID: PMC9944349 DOI: 10.3389/fimmu.2023.1127071] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) in the form of anti-CTLA-4 and anti-PD-1/PD-L1 have become the frontier of cancer treatment and successfully prolonged the survival of patients with advanced non-small cell lung cancer (NSCLC). But the efficacy varies among different patient population, and many patients succumb to disease progression after an initial response to ICIs. Current research highlights the heterogeneity of resistance mechanisms and the critical role of tumor microenvironment (TME) in ICIs resistance. In this review, we discussed the mechanisms of ICIs resistance in NSCLC, and proposed strategies to overcome resistance.
Collapse
Affiliation(s)
- Kexun Zhou
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, China
- Abdominal Oncology Ward, Division of Radiation Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shuo Li
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center, West China Hospital Sichuan University, Chengdu, China
| | - Yi Zhao
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Ke Cheng
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, China
- Abdominal Oncology Ward, Division of Radiation Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
44
|
Tomlins SA, Khazanov NA, Bulen BJ, Hovelson DH, Shreve MJ, Lamb LE, Matrana MR, Burkard ME, Yang ESH, Edenfield WJ, Dees EC, Onitilo AA, Thompson M, Buchschacher GL, Miller AM, Menter A, Parsons B, Wassenaar T, Hwang LC, Suga JM, Siegel R, Irvin W, Nair S, Slim JN, Misleh J, Khatri J, Masters G, Thomas S, Safa M, Anderson DM, Kwiatkowski K, Mitchell K, Hu-Seliger T, Drewery S, Fischer A, Plouffe K, Czuprenski E, Hipp J, Reeder T, Vakil H, Johnson DB, Rhodes DR. Development and validation of an integrative pan-solid tumor predictor of PD-1/PD-L1 blockade benefit. COMMUNICATIONS MEDICINE 2023; 3:14. [PMID: 36750617 PMCID: PMC9905474 DOI: 10.1038/s43856-023-00243-7] [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: 01/25/2022] [Accepted: 01/12/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Anti-PD-1 and PD-L1 (collectively PD-[L]1) therapies are approved for many advanced solid tumors. Biomarkers beyond PD-L1 immunohistochemistry, microsatellite instability, and tumor mutation burden (TMB) may improve benefit prediction. METHODS Using treatment data and genomic and transcriptomic tumor tissue profiling from an observational trial (NCT03061305), we developed Immunotherapy Response Score (IRS), a pan-tumor predictive model of PD-(L)1 benefit. IRS real-world progression free survival (rwPFS) and overall survival (OS) prediction was validated in an independent cohort of trial patients. RESULTS Here, by Cox modeling, we develop IRS-which combines TMB with CD274, PDCD1, ADAM12 and TOP2A quantitative expression-to predict pembrolizumab rwPFS (648 patients; 26 tumor types; IRS-High or -Low groups). In the 248 patient validation cohort (248 patients; 24 tumor types; non-pembrolizumab PD-[L]1 monotherapy treatment), median rwPFS and OS are significantly longer in IRS-High vs. IRS-Low patients (rwPFS adjusted hazard ratio [aHR] 0.52, p = 0.003; OS aHR 0.49, p = 0.005); TMB alone does not significantly predict PD-(L)1 rwPFS nor OS. In 146 patients treated with systemic therapy prior to pembrolizumab monotherapy, pembrolizumab rwPFS is only significantly longer than immediately preceding therapy rwPFS in IRS-High patients (interaction test p = 0.001). In propensity matched lung cancer patients treated with first-line pembrolizumab monotherapy or pembrolizumab+chemotherapy, monotherapy rwPFS is significantly shorter in IRS-Low patients, but is not significantly different in IRS-High patients. Across 24,463 molecularly-evaluable trial patients, 7.6% of patients outside of monotherapy PD-(L)1 approved tumor types are IRS-High/TMB-Low. CONCLUSIONS The validated, predictive, pan-tumor IRS model can expand PD-(L)1 monotherapy benefit outside currently approved indications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mark E Burkard
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Eddy Shih-Hsin Yang
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | | | - E Claire Dees
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| | - Adedayo A Onitilo
- Cancer Care and Research Center, Marshfield Clinic Research Institute, Marshfield, WI, USA
| | - Michael Thompson
- Aurora Cancer Care, Advocate Aurora Health, Milwaukee, WI, USA
- Tempus Labs, Chicago, IL, USA
| | | | - Alan M Miller
- SCL Health-CO, Broomfield, CO, USA
- Translational Drug Development, Scottsdale, USA
| | | | | | | | - Leon C Hwang
- Kaiser Permanente of the Mid-Atlantic States, Rockville, MD, USA
| | - J Marie Suga
- Kaiser Permanente Northern California, Vallejo, CA, USA
| | - Robert Siegel
- Bon Secours St. Francis Cancer Center, Greenville, SC, USA
| | | | - Suresh Nair
- Lehigh Valley Topper Cancer Institute, Allentown, PA, USA
| | | | | | - Jamil Khatri
- ChristianaCare Oncology Hematology, Newark, DE, USA
| | - Gregory Masters
- Medical Oncology Hematology Consultants, Helen F Graham Cancer Center and Research Institute,, Newark, DE, USA
| | - Sachdev Thomas
- Kaiser Permanente - Northern California, Oakland, CA, USA
| | | | - Daniel M Anderson
- Metro-Minnesota Community Oncology Research Consortium, St. Louis Park, MN, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
SMARCA4: Current status and future perspectives in non-small-cell lung cancer. Cancer Lett 2023; 554:216022. [PMID: 36450331 DOI: 10.1016/j.canlet.2022.216022] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/07/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022]
Abstract
SMARCA4, also known as transcription activator, is an ATP-dependent catalytic subunit of SWI/SNF (SWItch/Sucrose NonFermentable) chromatin-remodeling complexes that participates in the regulation of chromatin structure and gene expression by supplying energy. As a tumor suppressor that has aberrant expression in ∼10% of non-small-cell lung cancers (NSCLCs), SMARCA4 possesses many biological functions, including regulating gene expression, differentiation and transcription. Furthermore, NSCLC patients with SMARCA4 alterations have a weak response to conventional chemotherapy and poor prognosis. Therefore, the mechanisms of SMARCA4 in NSCLC development urgently need to be explored to identify novel biomarkers and precise therapeutic strategies for this subtype. This review systematically describes the biological functions of SMARCA4 and its role in NSCLC development, metastasis, functional epigenetics and potential therapeutic approaches for NSCLCs with SMARCA4 alterations. Additionally, this paper explores the relationship and regulatory mechanisms shared by SMARCA4 and its mutually exclusive catalytic subunit SMARCA2. We aim to provide innovative treatment strategies and improve clinical outcomes for NSCLC patients with SMARCA4 alterations.
Collapse
|
46
|
Di Federico A, De Giglio A, Gelsomino F, Sperandi F, Melotti B, Ardizzoni A. Predictors of survival to immunotherapy and chemoimmunotherapy in non-small cell lung cancer: A meta-analysis. J Natl Cancer Inst 2023; 115:29-42. [PMID: 36322815 DOI: 10.1093/jnci/djac205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/28/2022] [Accepted: 10/14/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Many patients with non-small cell lung cancer (NSCLC) derive poor benefit from immunotherapy (IO). For some of them, adding chemotherapy (CT) can improve the outcomes, but the reliability of programmed death-ligand 1 (PD-L1) expression as the only biomarker to distinguish these patients is unsatisfactory. We sought to detect clinicopathological and molecular predictive factors of survival that might be added to PD-L1 expression in the selection of patients who should receive IO alone or chemoimmunotherapy (CIT). METHODS We conducted a systematic search of randomized controlled clinical trials investigating IO, alone or with CT, vs CT alone in treatment-naïve advanced NSCLC patients. Meta-analyses and meta-regression analyses were performed to investigate IO alone vs CT, CIT vs CT, and IO alone vs CIT. RESULTS A total of 14 367 patients with advanced NSCLC across 25 randomized controlled clinical trials were included. Squamous histology, male sex, current and former smoker status, PD-L1 expression of 50% or more, and high tumor mutational burden (TMB) correlated with improved survival with IO alone compared with CT. Conversely, female sex, no smoking history, negative PD-L1 expression, and low TMB correlated with unsatisfactory outcomes with IO alone vs CT but not with CIT vs CT. CIT improved survival vs IO alone in female patients, never smokers, those having a PD-L1 expression of 1% or more (but not with a PD-L1 of ≥ 50%) or a low TMB and in patients with central nervous system metastasis. CONCLUSIONS These findings suggest some clinicopathological and molecular features that, added to PD-L1 expression, could help in the selection of the most appropriate first-line IO-based treatment for advanced NSCLC patients.
Collapse
Affiliation(s)
- Alessandro Di Federico
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Andrea De Giglio
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Francesco Gelsomino
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Francesca Sperandi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Barbara Melotti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrea Ardizzoni
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| |
Collapse
|
47
|
Rosen JC, Sacher A, Tsao MS. Direct GDP-KRAS G12C inhibitors and mechanisms of resistance: the tip of the iceberg. Ther Adv Med Oncol 2023; 15:17588359231160141. [PMID: 36950276 PMCID: PMC10026147 DOI: 10.1177/17588359231160141] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 02/06/2023] [Indexed: 03/18/2023] Open
Abstract
Kirsten rat sarcoma viral oncogene homolog mutations are observed in 25% of lung adenocarcinoma and 40% of these are G12C mutations. Historically, no approved targeted agents were available for patients with any KRAS mutation, and response rates to standard-of-care therapies were suboptimal. Newly developed inhibitors directed toward KRASG12C have been successful in clinical trials with overall response rates ranging between 32% and 46%, and two FDA approvals were granted in May 2021 and December 2022 as second-line or later monotherapies. However, rapid tumor resistance complicates their use as a monotherapy. With the rapid development of this novel class of inhibitors, it is important to discern the different types of tumor resistance that may arise and how each can differently contribute to tumor growth and survival. G12C inhibitor resistance is under investigation and combinations of therapies with G12C inhibitors have been proposed. Much of this insight is gleaned from preclinical investigations, as our knowledge of clinical resistance is in its infancy. In this review, we summarize the preclinical development of KRASG12C inhibitors, their clinical evaluations, different types of resistance mechanisms to these compounds, and ways of overcoming them. Finally, we underscore the importance of basic and translational investigations of these molecules in a landscape where their clinical evaluations garner the most attention, and we set the stage for what is to come.
Collapse
Affiliation(s)
- Joshua C. Rosen
- Princess Margaret Hospital Cancer Centre,
University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and
Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto,
ON, Canada
| | - Adrian Sacher
- Princess Margaret Hospital Cancer Centre,
University Health Network, Toronto, ON, Canada
- Division of Medical Oncology, Department of
Medicine, Princess Margaret Cancer Centre, Temerty Faculty of Medicine,
University of Toronto, Toronto, ON, Canada
- Department of Immunology, Temerty Faculty of
Medicine, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
48
|
Impact of Baseline Versus Intercurrent Steroids Administration on Upfront Chemo-Immunotherapy for Advanced Non-Small Cell Lung Cancer (NSCLC). Int J Mol Sci 2022; 23:ijms231810292. [PMID: 36142204 PMCID: PMC9499348 DOI: 10.3390/ijms231810292] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/30/2022] [Accepted: 09/06/2022] [Indexed: 12/02/2022] Open
Abstract
The impact of baseline versus intercurrent steroids on the efficacy of upfront chemotherapy plus pembrolizumab (CT-ICI) for advanced non-small cell lung cancer (NSCLC) patients is unclear. We conducted a retrospective study on metastatic NSCLC patients treated with upfront CT-ICI at our institution between March 2020 and December 2021. The use of steroids was considered as the administration of at least 10 mg of prednisone equivalent. Of 101 patients, 36 (35.6%) received steroid therapy at baseline, and 18 (17.8%) started steroids on treatment. Overall, median progression-free survival (mPFS) was 6.5 months (95% CI, 5.9−8.9) and median overall survival (mOS) was 18.2 months (95% CI, 8.9-NR). Patients taking baseline steroids had significantly shorter survival than those not taking them and those assuming intercurrent steroids (mPFS 5.0 vs. 9.2 vs. 7.3 months, p < 0.001; mOS 7.0 months vs. not reached, p < 0.001). Baseline steroids were significantly associated with poorer survival outcomes in the multivariate model (OS HR 2.94, p = 0.02; PFS HR 3.84, p > 0.001). Conversely, intercurrent prescription did not reach a significant value regardless of other pivotal variables included in the model. Baseline steroid administration was associated with a detrimental effect on survival outcomes in NSCLC patients treated with CT-ICI. The role of intercurrent steroid administration should be further explored in larger studies.
Collapse
|
49
|
Tokaz MC, Baik CS, Houghton AM, Tseng D. New Immuno-oncology Targets and Resistance Mechanisms. Curr Treat Options Oncol 2022; 23:1201-1218. [PMID: 35980521 DOI: 10.1007/s11864-022-01005-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2022] [Indexed: 11/28/2022]
Abstract
OPINION STATEMENT Immune checkpoint inhibition (ICI) has revolutionized the field of non-small cell lung cancer (NSCLC); currently, most patients with advanced disease receive upfront ICI either alone or in combination with chemotherapy. These advances have recently extended into early-stage NSCLC, with ICI incorporation into neoadjuvant and adjuvant treatment regimens. However, despite these successes, immunotherapy (IO) resistance remains a fundamental challenge in NSCLC, introducing a central quandary of how to precisely select the appropriate IO therapy or IO combination therapy for each individual patient. To address this vital need in the field, there has been an explosion of research in immuno-oncology to identify mechanisms of resistance, ranging from genomic alterations in the tumor to immunosuppressive conditions in the tumor microenvironment (TME). There remain many questions about how this complex interplay between the tumor and the immune microenvironment translates into clinical phenotypes of primary and acquired resistance. In NSCLC, a number of novel therapeutics are being developed to prevent and overcome resistance to ICI. Particular promise has been shown with therapeutics targeting novel T cell immune checkpoint inhibitors and targeting innate immune cells in the TME, chief among these cells are natural killer cells, neutrophils, and macrophages. Further research into tissue-based and non-invasive biomarkers that can be prospectively integrated into therapeutic trial design will be critical to advance the field's understanding of individual resistance patterns and enable the ultimate goal of precision immuno-oncology.
Collapse
Affiliation(s)
- Molly C Tokaz
- Division of Medical Oncology, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Christina S Baik
- Division of Medical Oncology, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - A McGarry Houghton
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.,Division of Pulmonology and Critical Care Medicine, University of Washington, Seattle, WA, USA.,Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Diane Tseng
- Division of Medical Oncology, University of Washington, Seattle, WA, USA. .,Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA. .,Fred Hutchinson Cancer Center, Mail Stop LG-540, 825 Eastlake Ave E., Seattle, WA, 98109, USA.
| |
Collapse
|
50
|
Di Federico A, De Giglio A, Gelsomino F, De Biase D, Giunchi F, Palladini A, Sperandi F, Melotti B, Ardizzoni A. Genomic Landscape, Clinical Features and Outcomes of Non-Small Cell Lung Cancer Patients Harboring BRAF Alterations of Distinct Functional Classes. Cancers (Basel) 2022; 14:3472. [PMID: 35884534 PMCID: PMC9319412 DOI: 10.3390/cancers14143472] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/15/2022] [Accepted: 07/15/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND In non-small cell lung cancer (NSCLC), BRAF class 1 alterations are effectively targeted by BRAF inhibitors. Conversely, targeted therapies have very low or absent activity in patients carrying class 2 and 3 alterations. The spectrum of BRAF alterations in NSCLC patients, and their accompanying clinical features, genomic landscape and treatment outcomes have been poorly reported. PATIENTS AND METHODS We identified BRAF alterations of defined functional class across different tumors through a systematic review. Then, we selected NSCLC patients carrying BRAF alterations, according to the systematic review, in the cBioPortal (cBioPortal cohort) to collect and analyze clinical, biomolecular and survival data. Finally, we identified NSCLC patients carrying BRAF non-V600 mutations enrolled in POPLAR and OAK trials (POPLAR/OAK cohort), extracting clinical and survival data for survival analyses. RESULTS 100 different BRAF non-V600 alterations were identified through the systematic review. In the cBioPortal cohort (n = 139), patients harboring class 2 and 3 alterations were more frequently smokers and had higher tumor mutational burden compared to those carrying class 1 alterations. The spectrum of most frequently co-altered genes was significantly different between BRAF alterations classes, including SETD2, STK11, POM121L12, MUC16, KEAP1, TERT, TP53 and other genes. In the POPLAR/OAK cohort, patients carrying non-V600 BRAF alterations were characterized by poor prognosis compared to BRAF wild-type patients. CONCLUSIONS Different classes of BRAF alterations confer distinctive clinical features, biomolecular signature and disease behavior to NSCLC patients. Non-V600 alterations are characterized by poor prognosis, but key gene co-alterations involved in cancer cell survival and immune pathways may suggest their potential sensitivity to tailored treatments.
Collapse
Affiliation(s)
- Alessandro Di Federico
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.D.G.); (F.G.); (F.S.); (B.M.); (A.A.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy;
| | - Andrea De Giglio
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.D.G.); (F.G.); (F.S.); (B.M.); (A.A.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy;
| | - Francesco Gelsomino
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.D.G.); (F.G.); (F.S.); (B.M.); (A.A.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy;
| | - Dario De Biase
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy;
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40138 Bologna, Italy
| | - Francesca Giunchi
- Pathology Department, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Arianna Palladini
- Department of Molecular Oncology, University of Pavia, 27100 Pavia, Italy;
| | - Francesca Sperandi
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.D.G.); (F.G.); (F.S.); (B.M.); (A.A.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy;
| | - Barbara Melotti
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.D.G.); (F.G.); (F.S.); (B.M.); (A.A.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy;
| | - Andrea Ardizzoni
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (A.D.G.); (F.G.); (F.S.); (B.M.); (A.A.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy;
| |
Collapse
|