1
|
Carcereny E, Rodriguez-Abreu D, Lopez R, Franco F, Guirado M, Massutí B, Cobo M, Blasco A, Suay G, Del Barco E, Ortega AL, Sala MA, Cordeiro P, Bernabé R, González Larriba JL, Bosch-Barrera J, Calzas J, Casal J, Padilla A, Sánchez-Hernandez A, Provencio M. Advanced non-squamous NSCLC with no actionable oncogenic driver in Spain: a cross-sectional descriptive analysis of data from the Thoracic Tumor Registry. Clin Transl Oncol 2024; 26:3218-3225. [PMID: 38862862 DOI: 10.1007/s12094-024-03511-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/26/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) accounts for the vast majority of all diagnosed lung cancers. According to their histology, most NSCLCs are considered non-squamous cell carcinoma (NSCC), and up to 85% of the latter may lack either one of the two main actionable oncogenic drivers (i.e., EGFR mutations and ALK rearrangements). OBJECTIVE Our analysis aimed to describe the clinical and epidemiological characteristics of Spanish patients suffering from NSCC with no actionable oncogenic driver in daily clinical practice. DESIGN A retrospective, cross-sectional, descriptive analysis. METHODS We analyzed the records of all Spanish patients with advanced NSCC diagnosed between January 2011 and January 2020 and included in the Spanish Thoracic Tumor Registry database. We evaluated the presence of metastasis and molecular profiling at the time of diagnosis and treatments received. We also assessed overall survival (OS) and progression-free survival (PFS) according to first-line treatment. RESULTS One thousand seven hundred ninety-seven Spanish patients with NSCC were included. They were mainly men (73.2%), smokers (current [44.4%] and former [44.4%]) and presented adenocarcinoma histology (97.6%). Most patients had at least one comorbidity (80.4%) and one metastatic site (96.8%), and a non-negligible number of those tested were PD-L1 positive (35.2%). Notably, the presence of liver metastasis indicated a shorter median OS and PFS than metastasis in other locations (p < 0.001). Chemotherapy was more often prescribed than immunotherapy as first-, second-, and third-line treatment in that period. In first-line, the OS rates were similar in patients receiving either regimen, but PFS rates significantly better in patients treated with immunotherapy (p = 0.026). Also, a high number of patients did not reach second- and third-line treatment, suggesting the failure of current early diagnostic measures and therapies. CONCLUSIONS This analysis of the most lethal tumor in Spain could highlight the strengths and the weaknesses of its clinical management and set the ground for further advances and research.
Collapse
Affiliation(s)
- Enric Carcereny
- Institut Català D'oncologia Badalona- Hospital Germans Trias I Pujol, B-Argo Group, Badalona, Spain.
| | | | - Rafael Lopez
- Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Fabio Franco
- Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Maria Guirado
- Hospital General Universitario de Elche, Elche, Spain
| | - Bartomeu Massutí
- Hospital General Universitario Dr. Balmis de Alicante, Alicante, Spain
| | - Manuel Cobo
- Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Ana Blasco
- Hospital General Universitario de Valencia, Valencia, Spain
| | - Guillermo Suay
- Hospital Universitari I Politècnic La Fe, Valencia, Spain
| | | | | | | | | | - Reyes Bernabé
- Hospital Universitario Virgen del Rocío, Seville, Spain
| | | | - Joaquim Bosch-Barrera
- Josep Trueta and Precision Oncology Group (OncoGIR-Pro), Institut d'Investigacions Biomèdiques de Girona (IDIBGI), Catalan Institute of Oncology, Hospital Universitari Dr, Girona, Spain
| | - Julia Calzas
- Hospital Universitario de Fuenlabrada, Fuenlabrada, Madrid, Spain
| | - Joaquín Casal
- Complejo Hospitalario Universitario de Vigo, Vigo, Spain
| | - Airam Padilla
- Hospital Universitario, Nuestra Señora De La Candelaria, Santa Cruz de Tenerife, Spain
| | | | | |
Collapse
|
2
|
Zheng C, Hu W, Wu D, Chen R, Xu C, Huang R. Foxd3/SLC5A6 axis regulates apoptosis in LUAD cells by controlling mitochondrial biotin uptake. Cell Signal 2024; 125:111473. [PMID: 39426496 DOI: 10.1016/j.cellsig.2024.111473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Lung cancer remains one of the leading causes of cancer-related mortality worldwide, with non-small cell lung cancer (NSCLC) accounting for over 85 % of cases. Lung adenocarcinoma (LUAD) is the most common subtype of NSCLC, and while targeted therapies and immune checkpoint inhibitors have improved outcomes, many patients exhibit resistance, necessitating the development of novel treatments. This study explores the role of the SLC5A6 gene, which encodes a sodium-dependent multivitamin transporter critical for mitochondrial function, in LUAD progression. We found that SLC5A6 is significantly upregulated in LUAD tissues and is associated with poor prognosis. Overexpression of SLC5A6 enhanced cell proliferation and migration, while knockout of SLC5A6 impaired these processes and induced apoptosis by disrupting mitochondrial function. Additionally, we identified Foxd3 as a key transcription factor regulating SLC5A6 expression. In vivo experiments demonstrated that SLC5A6 knockout effectively inhibited tumor growth. These findings suggest that SLC5A6 is a potential therapeutic target for LUAD, offering a new avenue for treatment strategies.
Collapse
Affiliation(s)
- Chong Zheng
- Department of Thoracic Surgery, The Dingli Clinical College of Wenzhou Medical University,Wenzhou Central Hospital, Wenzhou, China
| | - Wenxuan Hu
- Institute of Thoracic Surgery,The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Danni Wu
- Department of Thoracic Surgery, The Dingli Clinical College of Wenzhou Medical University,Wenzhou Central Hospital, Wenzhou, China
| | - Ruiheng Chen
- Department of Thoracic Surgery, The Dingli Clinical College of Wenzhou Medical University,Wenzhou Central Hospital, Wenzhou, China
| | - Chun Xu
- Institute of Thoracic Surgery,The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Risheng Huang
- Department of Thoracic Surgery, The Dingli Clinical College of Wenzhou Medical University,Wenzhou Central Hospital, Wenzhou, China.
| |
Collapse
|
3
|
Loo SK, Sica G, Wang X, Li T, Chen L, Gaither-Davis A, Huang Y, Burns TF, Stabile LP, Gao SJ. CASTOR1 phosphorylation predicts poor survival in male patients with KRAS-mutated lung adenocarcinoma. Cell Biosci 2024; 14:127. [PMID: 39385301 PMCID: PMC11465729 DOI: 10.1186/s13578-024-01307-4] [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: 08/21/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Lung cancer, a leading global cause of cancer-related mortality, necessitates enhanced prognostic markers for improved treatment outcomes. We have previously shown a tumor suppressive role of cytosolic arginine sensor for mTORC1 subunit 1 (CASTOR1), which is targeted for degradation upon phosphorylation at S14 (pCASTOR1) in multiple types of cancer. This study focuses on the predictive value of pCASTOR1 in lung adenocarcinoma (LUAD) patients with KRAS mutations. RESULTS Employing a newly developed pCASTOR1 specific antibody, we found that tumor cells exhibited significantly elevated pCASTOR1 scores compared to non-tumor cells (P < 0.05). Higher pCASTOR1 scores predicted poorer overall survival (OS) (HR = 3.3, P = 0.0008) and relapse-free survival (RFS) (HR = 3.0, P = 0.0035) in male patients with KRAS mutations. pCASTOR1 remained an independent predictor for OS (HR = 4.1, P = 0.0047) and RFS (HR = 3.5, P = 0.0342) after controlling for other factors. Notably, in early-stage LUAD, elevated pCASTOR1 scores were associated with significantly worse OS (HR = 3.3, P = 0.0176) and RFS (HR = 3.1, P = 0.0277) in male patients with KRAS mutations, akin to late-stage patients. CONCLUSION Elevated pCASTOR1 scores serve as biomarkers predicting poorer OS and RFS in male LUAD patients with KRAS mutations, offering potential clinical utility in optimizing treatment strategies for this subgroup.
Collapse
Affiliation(s)
- Suet Kee Loo
- Cancer Virology Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Gabriel Sica
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Presbyterian Hospital, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Xian Wang
- Cancer Virology Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tingting Li
- Cancer Virology Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Luping Chen
- Cancer Virology Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Autumn Gaither-Davis
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yufei Huang
- Cancer Virology Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Electrical and Computer Engineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy F Burns
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Cancer Biology Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Laura P Stabile
- Cancer Biology Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shou-Jiang Gao
- Cancer Virology Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
4
|
Jha P, Joshi A, Mishra R, Biswal RP, Kulkarni PM, Limaye S, Babu G, Batra U, Malik P, Kumar R, Shah M, Menon N, Rauthan A, Kuriakose M, Ramachandran V, Noronha V, Kumar P, Prabhash K. Landscape of Clinically Relevant Genomic Alterations in the Indian Non-small Cell Lung Cancer Patients. Clin Lung Cancer 2024:S1525-7304(24)00149-9. [PMID: 39129089 DOI: 10.1016/j.cllc.2024.07.011] [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: 05/28/2024] [Revised: 07/18/2024] [Accepted: 07/18/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND The genomic landscape of non-small cell lung cancer (NSCLC) in the Indian patients remains underexplored. We revealed distinctive genomic alterations of Indian NSCLC patients, thereby providing vital molecular insights for implementation of precision therapies. METHODS We analyzed the genomic profiles of 325 lung adenocarcinoma and 81 lung squamous carcinoma samples from Indian patients using targeted sequencing of 50 cancer related genes. Correlations between genomic alterations and clinical characteristics were computed using statistical analyses. Additionally, we identified distinct features of Indian NSCLC genomes by comparison across different ethnicities. RESULTS Our genomic analysis revealed several noticeable features of Indian NSCLC patients. Alterations in EGFR (45.8%), TP53 (27.4%), ALK (11.4%) and KRAS (10.2%) were predominant in adenocarcinoma, with 68% eligible for targeted therapies. Squamous carcinoma exhibited prevalent alterations in TP53 (40.7%), PIK3CA (17.3%), and CDKN2A (8.6%). We observed higher frequency of EGFR alterations (18.5%) in lung squamous carcinoma patients, significantly distinct from other ethnicities reported till date. Beyond established correlations, we observed 60% of PD-L1 negative squamous patients harbored TP53 alterations, suggesting intriguing therapeutic implications. CONCLUSIONS Our data revealed unique genomic variations of adenocarcinoma and squamous carcinoma patients, with significant indications for precision medicine and clinical practice of lung cancers. The study emphasizes the importance of clinical utility of NGS for routine diagnostics.
Collapse
Affiliation(s)
- Prerana Jha
- Department of Research, Karkinos Foundation, Mumbai, Maharashtra, India; Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India
| | - Asim Joshi
- Department of Research, Karkinos Foundation, Mumbai, Maharashtra, India; Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India
| | - Rohit Mishra
- Department of Research, Karkinos Foundation, Mumbai, Maharashtra, India; Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India
| | - Ranendra Pratap Biswal
- Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India
| | - Pooja Mahesh Kulkarni
- Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India
| | - Sewanti Limaye
- Department of Medical and Precision Oncology, Sir HN Reliance Foundation Hospital and Research Centre, Mumbai, Maharashtra, India
| | - Govind Babu
- Department of Medical Oncology, HCG Cancer Hospital, Bengaluru, India
| | - Ullas Batra
- Department of Medical Oncology, Section of Molecular Diagnostics, Pathology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Prabhat Malik
- Department of Medical Oncology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | - Rajiv Kumar
- Department of Pathology, Tata Memorial Centre, Mumbai, Maharashtra, India
| | - Minit Shah
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Nandini Menon
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Amit Rauthan
- Department of Medical Oncology, Manipal Hospital, Bengaluru, Karnataka, India
| | - Moni Kuriakose
- Department of Research, Karkinos Foundation, Mumbai, Maharashtra, India; Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India
| | - Venkataramanan Ramachandran
- Department of Research, Karkinos Foundation, Mumbai, Maharashtra, India; Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Prashant Kumar
- Department of Research, Karkinos Foundation, Mumbai, Maharashtra, India; Division of Cancer Biology, Karkinos Healthcare Pvt Ltd, Navi Mumbai, Maharashtra, India; Centre of Excellence for Cancer - Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh India.
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, Maharashtra, India.
| |
Collapse
|
5
|
Tian W, Niu L, Shi Y, Li S, Zhou R. First-line treatments for advanced non-squamous non-small cell lung cancer with immune checkpoint inhibitors plus chemotherapy: a systematic review, network meta-analysis, and cost-effectiveness analysis. Ther Adv Med Oncol 2024; 16:17588359241255613. [PMID: 38827178 PMCID: PMC11143870 DOI: 10.1177/17588359241255613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 04/30/2024] [Indexed: 06/04/2024] Open
Abstract
Introduction The combination of immune checkpoint inhibitors (ICIs) and chemotherapy is a promising first-line therapy for patients with advanced non-squamous non-small cell lung cancer (NSCLC). The cost-effectiveness of combinations with different ICIs is yet to be compared. Methods We utilized Bayesian network meta-analyses for the comparisons of overall survival, progression-free survival, and incidence of adverse events of the included treatments in the total population and subgroups with different programmed death-ligand 1 tumor proportional scores (TPS). The cost-effectiveness of the treatments from the perspectives of the US and Chinese healthcare systems was assessed using Markov models. Results Three combinations, including pembrolizumab + chemotherapy (PembroC), nivolumab + ipilimumab + chemotherapy (NivoIpiC), and atezolizumab + chemotherapy (AteC), were included in our study. In terms of efficacy, PembroC was most likely to be ranked first for extending progression-free survival (PFS) (93.16%) and overall survival (OS) (90.73%). Nevertheless, from the US perspective, NivoIpiC and PembroC showed incremental cost-effectiveness ratios (ICERs) of $68,963.1/quality-adjusted life-years (QALY) and $179,355.6/QALY, respectively, compared with AteC. The one-way sensitivity analysis revealed that the results were primarily sensitive to the hazard ratios for OS or the cost of immunotherapy agents. At a willingness-to-pay (WTP) threshold of $150,000/QALY, NivoIpiC had the highest probability of being cost-effective (63%). As for the Chinese perspective, NivoIpiC and PembroC had ICERs of $145,983.4/QALY and $195,863.3/QALY versus AteC, respectively. The results were primarily sensitive to the HRs for OS. At a WTP threshold of $38,017/QALY, AteC had the highest probability of cost-effectiveness (94%). Conclusion Although PembroC has the optimal efficacy, NivoIpiC and AteC were the most favorable treatments in terms of cost-effectiveness for patients with advanced non-squamous NSCLC from the US and Chinese perspectives, respectively.
Collapse
Affiliation(s)
- Wentao Tian
- Department of Oncology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan, China
| | - Lishui Niu
- Department of Oncology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan, China
| | - Yin Shi
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan 41008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan 410008, China
| | - Shuishi Li
- Department of General Surgery, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan, China
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan 410008, China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Road, Changsha, Hunan 410008, China
| |
Collapse
|
6
|
Kouchaki H, Kamyab P, Darbeheshti F, Gharezade A, Fouladseresht H, Tabrizi R. miR-939, as an important regulator in various cancers pathogenesis, has diagnostic, prognostic, and therapeutic values: a review. J Egypt Natl Canc Inst 2024; 36:16. [PMID: 38679648 DOI: 10.1186/s43046-024-00220-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/06/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND MicroRNAs (miRNAs or miRs) are highly conserved non-coding RNAs with a short length (18-24 nucleotides) that directly bind to a complementary sequence within 3'-untranslated regions of their target mRNAs and regulate gene expression, post-transcriptionally. They play crucial roles in diverse biological processes, including cell proliferation, apoptosis, and differentiation. In the context of cancer, miRNAs are key regulators of growth, angiogenesis, metastasis, and drug resistance. MAIN BODY This review primarily focuses on miR-939 and its expanding roles and target genes in cancer pathogenesis. It compiles findings from various investigations. MiRNAs, due to their dysregulated expression in tumor environments, hold potential as cancer biomarkers. Several studies have highlighted the dysregulation of miR-939 expression in human cancers. CONCLUSION Our study highlights the potential of miR-939 as a valuable target in cancer diagnosis, prognosis, and treatment. The aberrant expression of miR-939, along with other miRNAs, underscores their significance in advancing our understanding of cancer biology and their promise in personalized cancer care.
Collapse
Affiliation(s)
- Hosein Kouchaki
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parnia Kamyab
- USERN Office, Fasa University of Medical Sciences, Fasa, Iran
| | - Farzaneh Darbeheshti
- Department of Radiation Oncology, Dana Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Arezou Gharezade
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamed Fouladseresht
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Reza Tabrizi
- Clinical Research Development Unit, Valiasr Hospital, Fasa University of Medical Sciences, Fasa, Iran.
- Noncommunicable Diseases Research Center, Fasa University of Medical Science, Fasa, Iran.
| |
Collapse
|
7
|
Li Q, Song Q, Pei H, Chen Y. Emerging mechanisms of ferroptosis and its implications in lung cancer. Chin Med J (Engl) 2024; 137:818-829. [PMID: 38494343 PMCID: PMC10997236 DOI: 10.1097/cm9.0000000000003048] [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: 06/28/2023] [Indexed: 03/19/2024] Open
Abstract
ABSTRACT Lung cancer is one of the most common malignancies and has the highest number of deaths among all cancers. Despite continuous advances in medical strategies, the overall survival of lung cancer patients is still low, probably due to disease progression or drug resistance. Ferroptosis is an iron-dependent form of regulated cell death triggered by the lethal accumulation of lipid peroxides, and its dysregulation is implicated in cancer development. Preclinical evidence has shown that targeting the ferroptosis pathway could be a potential strategy for improving lung cancer treatment outcomes. In this review, we summarize the underlying mechanisms and regulatory networks of ferroptosis in lung cancer and highlight ferroptosis-targeting preclinical attempts to provide new insights for lung cancer treatment.
Collapse
Affiliation(s)
- Qian Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Huadong Pei
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington D.C. 20057, USA
| | - Yali Chen
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| |
Collapse
|
8
|
Khodabakhshi Z, Amini M, Hajianfar G, Oveisi M, Shiri I, Zaidi H. Dual-Centre Harmonised Multimodal Positron Emission Tomography/Computed Tomography Image Radiomic Features and Machine Learning Algorithms for Non-small Cell Lung Cancer Histopathological Subtype Phenotype Decoding. Clin Oncol (R Coll Radiol) 2023; 35:713-725. [PMID: 37599160 DOI: 10.1016/j.clon.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 06/10/2023] [Accepted: 08/05/2023] [Indexed: 08/22/2023]
Abstract
AIMS We aimed to build radiomic models for classifying non-small cell lung cancer (NSCLC) histopathological subtypes through a dual-centre dataset and comprehensively evaluate the effect of ComBat harmonisation on the performance of single- and multimodality radiomic models. MATERIALS AND METHODS A public dataset of NSCLC patients from two independent centres was used. Two image fusion methods, namely guided filtering-based fusion and image fusion based on visual saliency map and weighted least square optimisation, were used. Radiomic features were extracted from each scan, including first-order, texture and moment-invariant features. Subsequently, ComBat harmonisation was applied to the extracted features from computed tomography (CT), positron emission tomography (PET) and fused images to correct the centre effect. For feature selection, least absolute shrinkage and selection operator (Lasso) and recursive feature elimination (RFE) were investigated. For machine learning, logistic regression (LR), support vector machine (SVM) and AdaBoost were evaluated for classifying NSCLC subtypes. Training and evaluation of the models were carried out in a robust framework to offset plausible errors and performance was reported using area under the curve, balanced accuracy, sensitivity and specificity before and after harmonisation. N-way ANOVA was used to assess the effect of different factors on the performance of the models. RESULTS Support vector machine fed with selected features by recursive feature elimination from a harmonised PET feature set achieved the highest performance (area under the curve = 0.82) in classifying NSCLC histopathological subtypes. Although the performance of the models did not significantly improve for CT images after harmonisation, the performance of PET and guided filtering-based fusion feature signatures significantly improved for almost all models. Although the selection of the image modality and feature selection methods was effective on the performance of the model (ANOVA P-values <0.001), machine learning and harmonisation did not change the performance significantly (ANOVA P-values = 0.839 and 0.292, respectively). CONCLUSION This study confirmed the potential of radiomic analysis on PET, CT and hybrid images for histopathological classification of NSCLC subtypes.
Collapse
Affiliation(s)
- Z Khodabakhshi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
| | - M Amini
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - G Hajianfar
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - M Oveisi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran; Comprehensive Cancer Centre, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, Kings College London, London, UK; Department of Computer Science, University of British Columbia, Vancouver, BC, Canada
| | - I Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - H Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland; Geneva University Neurocenter, Geneva University, Geneva, Switzerland; Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
9
|
Chen W, Chen J, Zhang L, Cheng S, Yu J. Network meta-analysis of first-line immune checkpoint inhibitor therapy in advanced non-squamous non-small cell lung cancer patients with PD-L1 expression ≥ 50. BMC Cancer 2023; 23:791. [PMID: 37612622 PMCID: PMC10464425 DOI: 10.1186/s12885-023-11285-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/10/2023] [Indexed: 08/25/2023] Open
Abstract
INTRODUCTION The optimal first-line immunotherapy regimen for advanced non-squamous non-small cell lung cancer (NS-NSCLC) patients with programmed cell death ligand 1 (PD-L1) expression ≥ 50% remains unclear. Our aim is to determine the most effective treatment regimen through a network meta-analysis (NMA) comparing these treatments. METHODS A systematic search was performed in PubMed, Cochrane Library, Web of Science, and Embase databases, and a Bayesian network meta-analysis was conducted. To ensure transparency, the study was registered in the International Prospective Register of Systematic Reviews (CRD42022349712). RESULTS The analysis included 11 randomized controlled trials (RCTs) with 2037 patients and 12 immunotherapy combinations. ICI-ICI, ICI alone, and chemotherapy-ICI showed significant advantages over chemotherapy in terms of overall survival (OS) and progression-free survival (PFS). Pembrolizumab plus chemotherapy showed the best OS results compared to chemotherapy. Tislelizumab plus chemotherapy and sintilimab plus chemotherapy provided the best PFS results. CONCLUSIONS For NS-NSCLC patients with PD-L1 ≥ 50%, pembrolizumab plus chemotherapy, tislelizumab plus chemotherapy, and sintilimab plus chemotherapy are recommended as good treatment options based on the results of this Network meta-analysis (NMA).
Collapse
Affiliation(s)
- Wei Chen
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jiayi Chen
- School of Nursing, Capital Medical University, Beijing, China
| | - Lin Zhang
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Sheng Cheng
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Junxian Yu
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
10
|
Jiang S, Xiao M, Shi Y, Wang Y, Xu Z, Wang K. Identification of m7G-Related miRNA Signatures Associated with Prognosis, Oxidative Stress, and Immune Landscape in Lung Adenocarcinoma. Biomedicines 2023; 11:1569. [PMID: 37371664 DOI: 10.3390/biomedicines11061569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/13/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The role of N7-methylguanosine(m7G)-related miRNAs in lung adenocarcinoma (LUAD) remains unclear. We used LUAD data from The Cancer Genome Atlas (TCGA) to establish a risk model based on the m7G-related miRNAs, and divided patients into high-risk or low-risk subgroups. A nomogram for predicting overall survival (OS) was then constructed based on the independent risk factors. In addition, we performed a functional enrichment analysis and defined the oxidative stress-related genes, immune landscape as well as a drug response profile in the high-risk and low-risk subgroups. This study incorporated 28 m7G-related miRNAs into the risk model. The data showed a significant difference in the OS between the high-risk and low-risk subgroups. The receiver operating characteristic curve (ROC) predicted that the area under the curve (AUC) of one-year, three-year and five-year OS was 0.781, 0.804 and 0.853, respectively. The C-index of the prognostic nomogram for predicting OS was 0.739. We then analyzed the oxidative stress-related genes and immune landscape in the high-risk and low-risk subgroups. The data demonstrated significant differences in the expression of albumin (ALB), estimated score, immune score, stromal score, immune cell infiltration and functions between the high-risk and low-risk subgroups. In addition, the drug response analysis showed that low-risk subgroups may be more sensitive to tyrosine kinase inhibitor (TKI) and histone deacetylase (HDAC) inhibitors. We successfully developed a novel risk model based on m7G-related miRNAs in this study. The model can predict clinical prognosis and guide therapeutic regimens in patients with LUAD. Our data also provided new insights into the molecular mechanisms of m7G in LUAD.
Collapse
Affiliation(s)
- Sujing Jiang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Mingshu Xiao
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Yueli Shi
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Yongfang Wang
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Zhiyong Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China
| |
Collapse
|
11
|
Nandipati BL, Devarakonda N. Effective lung cancer diagnosis using multi-focus fusion of CT and PET images with deep learning strategies. THE IMAGING SCIENCE JOURNAL 2023. [DOI: 10.1080/13682199.2023.2183313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/01/2023]
|
12
|
Menon T, Gopal S, Rastogi Verma S. Targeted therapies in non-small cell lung cancer and the potential role of AI interventions in cancer treatment. Biotechnol Appl Biochem 2023; 70:344-356. [PMID: 35609005 DOI: 10.1002/bab.2356] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 04/17/2022] [Indexed: 11/12/2022]
Abstract
Non-small cell lung cancer is the most prevalent lung cancer, and almost three-fourths of patients are diagnosed in the advanced stage directly. In this stage, chemotherapy gives only a 15% 5-year survival rate. As people have varied symptoms and reactions to a specific cancer type, treatment for the tumor is likely to fall short, complicating cancer therapy. Immunotherapy is a breakthrough treatment involving drugs targeting novel immune checkpoint inhibitors like CTLA-4 and PD-1/PD-L1, along with combination therapies. In addition, the utility of engineered CAR-T and CAR-NK cells can be an effective strategy to promote the immune response against tumors. The concept of personalized cancer vaccines with the discovery of neoantigens loaded on dendritic cell vectors can also be an effective approach to cure cancer. Advances in genetic engineering tools like CRISPR/Cas9-mediated gene editing of T cells to enhance their effector function is another ray of hope. This review aims to provide an overview of recent developments in cancer immunotherapy, which can be used in first- and second-line treatments in the clinical space. Further, the intervention of artificial intelligence to detect cancer tumors at an initial stage with the help of machine learning techniques is also explored.
Collapse
Affiliation(s)
- Tarunya Menon
- Department of Biotechnology, Delhi Technological University, Delhi, India
| | - Shubhang Gopal
- Department of Information Technology, Delhi Technological University, Delhi, India
| | | |
Collapse
|
13
|
Natural history of lung squamous cell brain metastases in patients treated with radiosurgery: a thirty-year experience at a tertiary medical center. J Neurooncol 2023; 161:135-146. [PMID: 36469189 DOI: 10.1007/s11060-022-04153-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 09/29/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE In this study we report our 30-year experience in stereotactic radiosurgery (SRS) treatment of lung squamous cell carcinoma (LUSC) brain metastases (BMs). It will serve to provide detailed longitudinal outcomes and predictors of efficacy in treating LUSC-BMs with SRS. METHOD We retrospectively reviewed 51 patients and 109 tumors treated with SRS at our center between 1993 and 2022. Patient demographics, PDL1 genotype, immunotherapy use and mortality cause were recorded. Radiological and clinical outcomes were followed at 1-3-month intervals post-SRS. Cox-regression analysis and Kaplan-Meier survival curves were performed in statistical analysis. RESULTS We included 37 male and 14 female patients (median age 62.7 years at BM diagnosis). Median overall survival (OS) time was 6.9 months, 6-month OS rate was 62.1%, and Karnofsky performance scale (KPS) was the only independent predictor. Median time for local control maintenance was 7.6 months, 6-month local control rate was 69.1%, with TKI as the only independent predictor. Median time to distant failure was 5.13 months, 6-month distant failure rate was 51.1%, and factors with significant impact included gender (p = 0.002), presence of extracranial metastases (p < 0.001), use of immunotherapy(p < 0.001), PDL1 genotype (p = 0.034), and total intracranial metastases number (p = 0.008). However, no definitive benefits of immunotherapy were identified in patients with higher PDL1 mutational tumors. CONCLUSION In this study we defined the natural history of disease progression and outcomes in SRS-treated LUSC-BM patients. We also identified predictors of OS and tumor control among these patients. The findings of this study will serve as a guide when counseling these patients for SRS.
Collapse
|
14
|
CircPIM3 regulates taxol resistance in non-small cell lung cancer via miR-338-3p/TNFAIP8 axis. Anticancer Drugs 2023; 34:115-125. [PMID: 36539365 DOI: 10.1097/cad.0000000000001347] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Numerous work has revealed the involvement of circular RNA (circRNA) in regulating chemotherapy resistance. Here, we investigate circPIM3 role in taxol (Tax) resistance in non-small cell lung cancer (NSCLC). CircPIM3, microRNA (miR)-338-3p and tumor necrosis factor-alpha-induced protein-8 (TNFAIP8) expression were detected via quantitative real-time PCR, western blot or immunohistochemistry assay. Tax resistance was evaluated using cell counting kit-8, cell proliferation was measured by colony formation assay, cell cycle and apoptosis were examined via flow cytometry. The interplay between miR-338-3p and circPIM3 or TNFAIP8 was confirmed by dual-luciferase reporter assay. Finally, the effect of circPIM3 on Tax resistance in NSCLC in vivo was investigated by xenograft models. CircPIM3 and TNFAIP8 were upregulated in Tax-resistant NSCLC tissue and cell samples. Reducing circPIM3 expression inhibited Tax resistance, proliferation and induced cycle arrest and apoptosis in Tax-resistant NSCLC cells. Mechanically, circPIM3 absence led to downregulation of TNFAIP8 via absorbing miR-338-3p. Additionally, circPIM3 depletion increased Tax sensitivity of NSCLC in vivo. Silencing of circPIM3 suppressed Tax resistance in Tax-resistant NSCLC cells through regulation of the miR-338-3p/TNFAIP8 axis.
Collapse
|
15
|
Radiosensitization-Related Cuproptosis LncRNA Signature in Non-Small Cell Lung Cancer. Genes (Basel) 2022; 13:genes13112080. [PMID: 36360316 PMCID: PMC9690519 DOI: 10.3390/genes13112080] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/28/2022] [Accepted: 11/01/2022] [Indexed: 11/12/2022] Open
Abstract
A new treatment modality targeting cuproptosis is gradually entering the public horizon. Cuproptosis is a new form of regulated cell death distinct from ferroptosis, apoptosis, autophagy, and necrosis. Previous studies have discovered that the copper level varies considerably in various cancers and that an increase in copper content is directly associated with the proliferation and metastasis of cancer cells. In non-small cell lung cancer (NSCLC) after radiation, the potential utility of cuproptosis-related long noncoding RNAs (lncRNAs) is still unclear. This research aimed to develop a prediction signature based on lncRNAs associated with cuproptosis to predict the prognosis of NSCLC patients following radiation. Methods: Expression data of primary tumors and adjacent solid tissues were downloaded from The Cancer Genome Atlas (TCGA) database, along with the corresponding clinical and mutational data. Univariate and multivariate COX analyses and LASSO regression analyses were performed to obtain a predictive signature of lncRNAs associated with cuproptosis. The data were randomly grouped into a training group used for model construction and a test group used for model validation. The model was validated by drawing a survival curve, risk curve, independent prognostic analysis, ROC curve PFS analysis, etc. Results: The lncRNA signature consisting of six cuproptosis-related lncRNAs (AC104088.1, PPP4R3B-DT, AC006042.3, LUCAT1, HHLA3-AS1, and LINC02029) was used to predict the prognosis of patients. Among them, there were three high-risk lncRNAs (LUCAT1, HHLA3-AS1, and LINC02029) with HR > 1 and three protective lncRNAs (AC104088.1, PPP4R3B-DT, and AC006042.3), with an HR < 1. Data analysis demonstrated that the cuproptosis-related lncRNA signatures could well predict the prognosis of NSCLC patients after radiation. Patients in the high-risk category receive a worse prognosis than those in the low-risk group. Cuproptosis-related risk prediction demonstrated better predictive qualities than age, gender, and pathological stage factors. Conclusion: The risk proposed model can independently predict the prognosis of NSCLC patients after radiotherapy, provide a foundation for the role of cuproptosis-related lncRNAs in NSCLC after radiotherapy, and provide a clinical strategy for radiotherapy combined with cuproptosis in NSCLC patients.
Collapse
|
16
|
Shao T, Zhao M, Liang L, Tang W. A systematic review and network meta-analysis of first-line immune checkpoint inhibitor combination therapies in patients with advanced non-squamous non-small cell lung cancer. Front Immunol 2022; 13:948597. [PMID: 36389713 PMCID: PMC9645411 DOI: 10.3389/fimmu.2022.948597] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/26/2022] [Indexed: 06/28/2024] Open
Abstract
INTRODUCTION Clinical evidence suggests that first-line immune checkpoint inhibitor (ICI) combination therapies can improve survival in patients with advanced non-squamous non-small cell lung cancer (nsq-NSCLC). However, the optimal strategy remains unknown without a systematic comparison of their long-term effects. METHODS We performed a systematic review and network meta-analysis by retrieving up-to-date literature from PubMed® (National Library of Medicine, Bethesda, MD, USA), Embase® (Elsevier, Amsterdam, Netherlands), MEDLINE® (National Library of Medicine), ClinicalTrials.gov (National Library of Medicine), and major international conference publications. Published studies and abstracts comparing first-line ICI combination therapies with other treatments for patients with advanced nsq-NSCLC were included. Restricted mean survival time (RMST) was measured over 12 months for progression-free survival (PFS) and 18 months for overall survival (OS), and the Royston-Parmar model was used to extrapolate and compare data for the long-term outcomes. RESULTS We included a total of 11 trials involving 12 therapies and 6,130 patients. Pembrolizumab plus chemotherapy exhibited the best overall survival (OS) benefit at both 18 and 60 months [RMST = 2.95, 95% confidence interval (CI) 1.96 to 3.97; life-years gained over a 5-year period = 2.18 years]. Nivolumab plus bevacizumab plus chemotherapy was found to present the best progression-free survival (PFS) benefit at 12 months (RMST 3.02, 95% CI 2.11 to 3.91), whereas atezolizumab plus bevacizumab plus chemotherapy showed the best PFS benefit at 36 months (life-years gained over 3 years = 1.22 years). Subgroup analyses showed that among patients with programmed death-ligand 1 (PD-L1) expression ≥ 50%, atezolizumab plus chemotherapy and nivolumab plus ipilimumab resulted in superior OS benefits at 18 and 60 months, respectively. Among patients with PD-L1 expression< 1%, pembrolizumab plus chemotherapy was associated with OS benefits at both 18 and 60 months. Sintilimab plus chemotherapy was associated with relatively fewer grade ≥ 3 adverse events than other ICI combination therapies. CONCLUSION Our results show that ICI combination therapies showed better survival benefits than chemotherapy. Pembrolizumab plus chemotherapy could provide the best OS benefits to patients with advanced nsq-NSCLC, whereas atezolizumab plus bevacizumab plus chemotherapy could bring the best PFS benefits. The optimal ICI combination therapy varies depending on PD-L1 expression level. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=325005, identifier CRD42022325005.
Collapse
Affiliation(s)
- Taihang Shao
- Center for Pharmacoeconomics and Outcomes Research, China Pharmaceutical University, Nanjing, China
| | - Mingye Zhao
- Center for Pharmacoeconomics and Outcomes Research, China Pharmaceutical University, Nanjing, China
| | - Leyi Liang
- Center for Pharmacoeconomics and Outcomes Research, China Pharmaceutical University, Nanjing, China
| | - Wenxi Tang
- Center for Pharmacoeconomics and Outcomes Research, China Pharmaceutical University, Nanjing, China
- Department of Public Affairs Management, School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
17
|
Zhu J, Pan F, Cai H, Pan L, Li Y, Li L, Li Y, Wu X, Fan H. Positron emission tomography imaging of lung cancer: An overview of alternative positron emission tomography tracers beyond F18 fluorodeoxyglucose. Front Med (Lausanne) 2022; 9:945602. [PMID: 36275809 PMCID: PMC9581209 DOI: 10.3389/fmed.2022.945602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
Lung cancer has been the leading cause of cancer-related mortality in China in recent decades. Positron emission tomography-computer tomography (PET/CT) has been established in the diagnosis of lung cancer. 18F-FDG is the most widely used PET tracer in foci diagnosis, tumor staging, treatment planning, and prognosis assessment by monitoring abnormally exuberant glucose metabolism in tumors. However, with the increasing knowledge on tumor heterogeneity and biological characteristics in lung cancer, a variety of novel radiotracers beyond 18F-FDG for PET imaging have been developed. For example, PET tracers that target cellular proliferation, amino acid metabolism and transportation, tumor hypoxia, angiogenesis, pulmonary NETs and other targets, such as tyrosine kinases and cancer-associated fibroblasts, have been reported, evaluated in animal models or under clinical investigations in recent years and play increasing roles in lung cancer diagnosis. Thus, we perform a comprehensive literature review of the radiopharmaceuticals and recent progress in PET tracers for the study of lung cancer biological characteristics beyond glucose metabolism.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China,Respiratory and Critical Care Medicine, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China,NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Fei Pan
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Huawei Cai
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lili Pan
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yalun Li
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Li
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - YunChun Li
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China,Department of Nuclear Medicine, The Second People’s Hospital of Yibin, Yibin, China
| | - Xiaoai Wu
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China,Xiaoai Wu,
| | - Hong Fan
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Hong Fan,
| |
Collapse
|
18
|
ABALI H, MANDAL ZİREK T, TOKGÖZ AKYIL F, ÖZBOLAT ZB, KOCAOĞLU A, KILINÇARSLAN KG, TURAL ÖNÜR S. Non-metastatic Squamous Cell Lung Carcinoma: A rare case report. IZMIR DEMOCRACY UNIVERSITY HEALTH SCIENCES JOURNAL 2022; 5:106-112. [DOI: 10.52538/iduhes.1060306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Central and peripheral types of squamous cell carcinoma (SQCC) are classified in different categories due to different clinical-pathological features. Peripheral SQCC, according to histological pattern; it is classified into three subgroups as alveolar space-filling type, expanding type and combined type. Of these three subtypes, alveolar space-filling neither causes lymphatic vessel invasion nor lymph node metastasis and has the best prognosis. Herein, we present a rare case of squamous cell carcinoma that did not metastasize at two and a half years years from initial diagnosis.
Collapse
Affiliation(s)
- Hülya ABALI
- UNIVERSITY OF HEALTH SCIENCES, İSTANBUL YEDİKULE HEALTH RESEARCH CENTER FOR PULMONOLOGY AND THORACIC SURGERY, DEPARTMENT OF INTERNAL MEDICINE
| | - Tugba MANDAL ZİREK
- UNIVERSITY OF HEALTH SCIENCES, İSTANBUL YEDİKULE HEALTH RESEARCH CENTER FOR PULMONOLOGY AND THORACIC SURGERY, DEPARTMENT OF INTERNAL MEDICINE
| | - Fatma TOKGÖZ AKYIL
- UNIVERSITY OF HEALTH SCIENCES, İSTANBUL YEDİKULE HEALTH RESEARCH CENTER FOR PULMONOLOGY AND THORACIC SURGERY, DEPARTMENT OF INTERNAL MEDICINE
| | - Zehra Büşra ÖZBOLAT
- UNIVERSITY OF HEALTH SCIENCES, İSTANBUL YEDİKULE HEALTH RESEARCH CENTER FOR PULMONOLOGY AND THORACIC SURGERY, DEPARTMENT OF INTERNAL MEDICINE
| | - Aslı KOCAOĞLU
- UNIVERSITY OF HEALTH SCIENCES, İSTANBUL YEDİKULE HEALTH RESEARCH CENTER FOR PULMONOLOGY AND THORACIC SURGERY, DEPARTMENT OF INTERNAL MEDICINE
| | - Kübra Gül KILINÇARSLAN
- UNIVERSITY OF HEALTH SCIENCES, İSTANBUL YEDİKULE HEALTH RESEARCH CENTER FOR PULMONOLOGY AND THORACIC SURGERY, DEPARTMENT OF INTERNAL MEDICINE
| | - Seda TURAL ÖNÜR
- UNIVERSITY OF HEALTH SCIENCES, İSTANBUL YEDİKULE HEALTH RESEARCH CENTER FOR PULMONOLOGY AND THORACIC SURGERY, DEPARTMENT OF INTERNAL MEDICINE
| |
Collapse
|
19
|
Ekinci M, Santos-Oliveira R, Ilem-Ozdemir D. Biodistribution of 99mTc-PLA/PVA/Atezolizumab Nanoparticles for Non-Small Cell Lung Cancer Diagnosis. Eur J Pharm Biopharm 2022; 176:21-31. [PMID: 35568255 DOI: 10.1016/j.ejpb.2022.05.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/13/2022] [Accepted: 05/09/2022] [Indexed: 01/17/2023]
Abstract
Lung cancer (LC) is a common type of cancer, which is a leading cause of death around the world. There is an urgency for the development of new drugs that could diagnose the LC in the early stages and in a precise manner. In this direction, the development of nanoparticles radiolabeled with the diagnostic radioisotopes represent an important advance in the field of cancer imaging. In this study were developed PLA/PVA/Atezolizumab nanoparticles which were radiolabeled with 99mTc (Technetium-99m). The radiolabeled nanoparticles were evaluated in both: in-vitro (L-929 and A-549) as in-vivo (mice). The results showed no cytotoxicity effect in the healthy cells (L-929) and cytotoxicity effect in the tumor cells (A-549). The biodistribution assay demonstrated that 99mTc-PLA/PVA/Atezolizumab could reach the tumor site 14-folds higher than the nonparticulate atezolizumab. In conclusion, 99mTc-PLA/PVA/Atezolizumab nanoparticles showed to be a new drug which is able to precisely image the lung tumor, and it must be considered for clinical trials.
Collapse
Affiliation(s)
- Meliha Ekinci
- Ege University, Faculty of Pharmacy, Department of Radiopharmacy, 35040 Bornova, Izmir, Turkey
| | - Ralph Santos-Oliveira
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Laboratory of Synthesis of Novel Radiopharmaceuticals and Nanoradiopharmacy, Rio de Janeiro, 21941906, Brazil; Zona Oeste State University, Laboratory of Nanoradiopharmaceuticals and Radiopharmacy, Rio de Janeiro, 23070200, Brazil
| | - Derya Ilem-Ozdemir
- Ege University, Faculty of Pharmacy, Department of Radiopharmacy, 35040 Bornova, Izmir, Turkey.
| |
Collapse
|
20
|
Lei Y, Wang K, Liu Y, Wang X, Xiang X, Ning X, Ding W, Duan J, Li D, Zhao W, Li Y, Zhang F, Luo X, Shi Y, Wang Y, Huang D, Bai Y, Zhang H. Various Subtypes of EGFR Mutations in Patients With NSCLC Define Genetic, Immunologic Diversity and Possess Different Prognostic Biomarkers. Front Immunol 2022; 13:811601. [PMID: 35265073 PMCID: PMC8899028 DOI: 10.3389/fimmu.2022.811601] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Based on data analysis of 9649 Chinese primary NSCLC patients, we calculated the exact proportion of EGFR subtypes in NSCLC and evaluated the TMB level, PD-L1 expression level and tumor immune microenvironment among different EGFR mutation subtypes. Postoperative follow-up data for 98 patients were collected and analyzed. The results showed that several uncommon EGFR mutation subtypes have a higher proportion of TMB-high or strong positive PD-L1 expression than the total EGFR mutation group. In addition, different subtypes have different characteristics related to the immune microenvironment, such as G719 mutations being associated with more CD8+ T cell infiltration into tumors; except for EGFR 19del, CD8+ T cell infiltration into tumors of other EGFR mutation subtypes were similar to that of wildtype EGFR. Moreover, follow-up results revealed that components of the immune microenvironment have prognostic value for NSCLC patients, with different prognostic biomarkers for NSCLC patients with and without EGFR mutations. These results suggest that patients with different EGFR mutations need to be treated differently. The prognosis of NSCLC patients may be assessed through components of tumor immune microenvironment, and ICIs treatment may be considered for those with some uncommon EGFR mutation subtypes.
Collapse
Affiliation(s)
- Youming Lei
- Department of Geriatric Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Kun Wang
- Department of Thoracic Surgery, Anning First Peoples Hospital affiliate to Kunming University of Science and Technology (Kunming Forth People's Hospital), Kunming, China
| | - Yinqiang Liu
- Department of Geriatric Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xuming Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xudong Xiang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiangu Ning
- Department of Thoracic Surgery, The First Peoples Hospital of Yunnan Province, Kunming, China
| | - Wanbao Ding
- Department of Oncology, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Jin Duan
- Department of Geriatric Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Dingbiao Li
- Department of Thoracic Surgery, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Wei Zhao
- Department of Geriatric Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yi Li
- Department of Oncology, Yunnan Provincial Hospital of Traditional Chinese Medicine, Kunming, China
| | - Fujun Zhang
- Department of Geriatric Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiaoyu Luo
- Department of Geriatric Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunfei Shi
- Department of Geriatric Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ying Wang
- Department of Thoracic Surgery, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, China
| | - Depei Huang
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Yuezong Bai
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Hushan Zhang
- The Medical Department, 3D Medicines Inc., Shanghai, China
| |
Collapse
|
21
|
Circ_0074027 regulates the progression of non-small cell lung cancer via miR-1304-5p/GJB2 axis. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00235-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
22
|
Seguin L, Durandy M, Feral CC. Lung Adenocarcinoma Tumor Origin: A Guide for Personalized Medicine. Cancers (Basel) 2022; 14:cancers14071759. [PMID: 35406531 PMCID: PMC8996976 DOI: 10.3390/cancers14071759] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 12/29/2022] Open
Abstract
Simple Summary Lung cancer is the leading cause of cancer-related death worldwide, with an average 5-year survival rate of approximately 15%. Among the multiple histological type of lung cancer, adenocarcinoma is the most common. Adenocarcinoma is characterized by a high degree of heterogeneity at many levels, including histological, cellular, and molecular. Understanding the cell of origin of adenocarcinoma, and the molecular changes during tumor progression, will allow better therapeutic strategies. Abstract Lung adenocarcinoma, the major form of lung cancer, is the deadliest cancer worldwide, due to its late diagnosis and its high heterogeneity. Indeed, lung adenocarcinoma exhibits pronounced inter- and intra-tumor heterogeneity cofounding precision medicine. Tumor heterogeneity is a clinical challenge driving tumor progression and drug resistance. Several key pieces of evidence demonstrated that lung adenocarcinoma results from the transformation of progenitor cells that accumulate genetic abnormalities. Thus, a better understanding of the cell of origin of lung adenocarcinoma represents an opportunity to unveil new therapeutic alternatives and stratify patient tumors. While the lung is remarkably quiescent during homeostasis, it presents an extensive ability to respond to injury and regenerate lost or damaged cells. As the lung is constantly exposed to potential insult, its regenerative potential is assured by several stem and progenitor cells. These can be induced to proliferate in response to injury as well as differentiate into multiple cell types. A better understanding of how genetic alterations and perturbed microenvironments impact progenitor-mediated tumorigenesis and treatment response is of the utmost importance to develop new therapeutic opportunities.
Collapse
|
23
|
Liang Z, Zhang E, Duan L, Weygant N, An G, Hu B, Yao J. Establishment of a Competing Risk Nomogram in Patients with Pulmonary Sarcomatoid Carcinoma. Technol Cancer Res Treat 2022; 21:15330338211068960. [PMID: 35179409 PMCID: PMC8859694 DOI: 10.1177/15330338211068960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background and aim: Pulmonary sarcomatoid carcinoma (PSC) is a rare subtype of nonsmall cell lung cancer with a poor prognosis. This study aimed to analyze the clinicopathological characteristics and survival outcomes among patients with PSC, lung squamous cell cancer (SCC), and lung adenocarcinoma (LAC), and to construct a competing risk nomogram for patients with PSC. Method: Data of 3 groups of patients diagnosed with PSC, SCC, or LAC from the surveillance, epidemiology, and end results (SEER) database between 1988 and 2015 were retrospectively reviewed. A 1:1 propensity score matching (PSM) analysis was used to balance the baseline data of patients. Independent risk factors associated with survival outcomes were screened by the least absolute shrinkage and selection operator and further determined by univariate and multivariate Cox proportional risk regression analyses. The overall survival (OS) of patients was evaluated by Kaplan–Meier analysis and compared with a log-rank test. The cumulative incidence function was used to estimate the 5-year probabilities of the cancer-specific mortality of PSC. A nomogram was constructed to illustrate the competing risk model to predict the 3- and 5-year OS, and corresponding concordance indexes (C-indexes) and calibration curves were used to assess and validate the competing risk nomogram. Results: A total of 2285 patients with PSC were included in this study. Compared with SCC and LAC patients, the Kaplan–Meier analysis showed that patients with PSC had a worse prognosis, with a median survival of 5 months (95% confidence interval [CI]: 5-6 months) and a 5-year OS rate of 15.3% (95% CI: 13.9%-16.9%). Similar outcomes were demonstrated after 1:1 PSM. Moreover, the competing risk model showed that age, T stage, M stage, tumor size, lymph node ratio (LNR), surgery, and chemotherapy were associated with PSC-specific mortality. The 5-year C-index of the nomogram was 0.718. Calibration curves illustrated that the nomogram was well-validated and had great accuracy. Conclusions: Patients with PSC had a worse survival outcome compared with SCC or LAC patients. Age, T stage, M stage, tumor size, LNR, surgery, and chemotherapy were associated with PSC-specific mortality. The competing risk nomogram displayed excellent discrimination in predicting PSC-specific mortality.
Collapse
Affiliation(s)
- Ziwei Liang
- Beijing Chao-Yang Hospital, 74639Capital Medical University, Beijing, China
| | - Enyu Zhang
- 71043Beijing Fu Xing Hospital, Capital Medical University, Beijing, China
| | - Ling Duan
- Beijing Chao-Yang Hospital, 74639Capital Medical University, Beijing, China
| | - Nathaniel Weygant
- 47858Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China.,Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, Fujian, China
| | - Guangyu An
- Beijing Chao-Yang Hospital, 74639Capital Medical University, Beijing, China
| | - Bin Hu
- Department of Thoracic Surgery, Beijing Chao-Yang Hospital, 74639Capital Medical University, Beijing, China
| | - Jiannan Yao
- Beijing Chao-Yang Hospital, 74639Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Modulation of chemoimmunotherapy efficacy in non-small cell lung cancer by sex and histology: a real-world, patient-level analysis. BMC Cancer 2022; 22:80. [PMID: 35045806 PMCID: PMC8767728 DOI: 10.1186/s12885-022-09187-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/20/2021] [Indexed: 12/14/2022] Open
Abstract
Background It has been postulated that patient’s sex impacts response to immunotherapy. Sex modulation of immunotherapy benefit, however, has not yet been explored using patient-level data, where potential confounders, as well as histologic type, can be accounted for. Here we investigated the association between sex and chemoimmunotherapy efficacy for non-small cell lung cancer (NSCLC) using a large, nation-wide dataset. Patients & methods Stage IV NSCLC patients diagnosed in 2015 were identified in the National Cancer Database (NCDB). Patients were treated with either chemoimmunotherapy or chemotherapy alone. The efficacy of the addition of immunotherapy treatment by sex was investigated using both an adjusted Cox proportional hazards model and propensity-score matching, in both the overall cohort and stratified by histological subtype. Results 2064 (16%) patients received chemoimmunotherapy and10,733 (84%) received chemotherapy alone. Adjusted survival analysis in the overall cohort showed that both males (hazards ratio (HR)adj: 0.80, 95% CI: 0.74–0.87) and females (HRadj: 0.83, 95% CI: 0.76–0.90) had better OS when treated with chemoimmunotherapy than chemotherapy alone, with no statistically significant interaction between sex and receipt of immunotherapy (p = 0.63). Propensity matching confirmed these results. However, for those with squamous cell histology, male patients derived more benefit from chemoimmunotherapy treatment than females (HRadj: 0.73, 95% CI: 0.58–0.91 vs HRadj: 1.03, 95% CI: 0.76–1.38; p for interaction = 0.07). Conclusion Male patients with squamous cell carcinoma may derive more benefit from chemoimmunotherapy treatment. Histology likely plays an important role in how sex modulates immunotherapy efficacy. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09187-y.
Collapse
|
25
|
PET imaging of lung and pleural cancer. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00206-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
26
|
Federico L, McGrail DJ, Bentebibel SE, Haymaker C, Ravelli A, Forget MA, Karpinets T, Jiang P, Reuben A, Negrao MV, Li J, Khairullah R, Zhang J, Weissferdt A, Vaporciyan AA, Antonoff MB, Walsh G, Lin SY, Futreal A, Wistuba I, Roth J, Byers LA, Gaudreau PO, Uraoka N, Cruz AF, Dejima H, Lazcano RN, Solis LM, Parra ER, Lee JJ, Swisher S, Cascone T, Heymach JV, Zhang J, Sepesi B, Gibbons DL, Bernatchez C. Distinct tumor-infiltrating lymphocyte landscapes are associated with clinical outcomes in localized non-small-cell lung cancer. Ann Oncol 2022; 33:42-56. [PMID: 34653632 PMCID: PMC10019222 DOI: 10.1016/j.annonc.2021.09.021] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 09/11/2021] [Accepted: 09/30/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Despite the importance of tumor-infiltrating T lymphocytes (TILs) in cancer biology, the relationship between TIL phenotypes and their prognostic relevance for localized non-small-cell lung cancer (NSCLC) has not been well established. PATIENTS AND METHODS Fresh tumor and normal adjacent tissue was prospectively collected from 150 patients with localized NSCLC. Tissue was comprehensively characterized by high-dimensional flow cytometry of TILs integrated with immunogenomic data from multiplex immunofluorescence, T-cell receptor sequencing, exome sequencing, RNA sequencing, targeted proteomics, and clinicopathologic features. RESULTS While neither the magnitude of TIL infiltration nor specific TIL subsets were significantly prognostic alone, the integration of high-dimensional flow cytometry data identified two major immunotypes (IM1 and IM2) that were predictive of recurrence-free survival independent of clinical characteristics. IM2 was associated with poor prognosis and characterized by the presence of proliferating TILs expressing cluster of differentiation 103, programmed cell death protein 1, T-cell immunoglobulin and mucin-domain containing protein 3, and inducible T-cell costimulator. Conversely, IM1 was associated with good prognosis and differentiated by an abundance of CD8+ T cells expressing cytolytic enzymes, CD4+ T cells lacking the expression of inhibitory receptors, and increased levels of B-cell infiltrates and tertiary lymphoid structures. While increased B-cell infiltration was associated with good prognosis, the best prognosis was observed in patients with tumors exhibiting high levels of both B cells and T cells. These findings were validated in patient tumors from The Cancer Genome Atlas. CONCLUSIONS Our study suggests that although the number of infiltrating T cells is not associated with patient survival, the nature of the infiltrating T cells, resolved in distinct TIL immunotypes, is prognostically relevant in NSCLC and may inform therapeutic approaches to clinical care.
Collapse
Affiliation(s)
- L Federico
- Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - D J McGrail
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - S-E Bentebibel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - C Haymaker
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - A Ravelli
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - M-A Forget
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - T Karpinets
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - P Jiang
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - A Reuben
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - M V Negrao
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Li
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - R Khairullah
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - A Weissferdt
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - A A Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - M B Antonoff
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - G Walsh
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - S-Y Lin
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - A Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Roth
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - L A Byers
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - P-O Gaudreau
- Department of Oncology, Queens' University and the Canadian Cancer Trials Group, Kingston, Canada
| | - N Uraoka
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - A F Cruz
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - H Dejima
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - R N Lazcano
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - L M Solis
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - E R Parra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J J Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - S Swisher
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - T Cascone
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA; Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA.
| | - B Sepesi
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, USA.
| | - D L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA.
| | - C Bernatchez
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA.
| |
Collapse
|
27
|
Sotelo MJ, Luis García J, Torres-Mattos C, Milián H, Carracedo C, González-Ruiz MÁ, Mielgo-Rubio X, Trujillo-Reyes JC, Couñago F. Recent advances and new insights in the management of early-stage epidermal growth factor receptor-mutated non-small-cell lung cancer. World J Clin Oncol 2021; 12:912-925. [PMID: 34733613 PMCID: PMC8546659 DOI: 10.5306/wjco.v12.i10.912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/29/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
Patients with early-stage non-small-cell lung cancer (NSCLC) are candidates for curative surgery; however, despite multiple advances in lung cancer management, recurrence rates remain high. Adjuvant chemotherapy has been demonstrated to significantly prolong overall survival (OS), but this benefit is modest and there is an urgent need for effective new therapies to provide a cure for more patients. The high efficacy of tyrosine kinase inhibitors (TKIs) against epidermal growth factor receptor-mutated (EGFR) in patients with advanced EGFR-mutated NSCLC has led to the evaluation of these agents in early stages of the disease. Multiple clinical trials have evaluated the safety and efficacy of EGFR TKIs as an adjuvant treatment, in patients with resected EGFR-mutated NSCLC, and shown that they significantly prolong disease-free survival (DFS), but this benefit does not translate to OS. Recently, an interim analysis of the ADAURA trial demonstrated that, surprisingly, osimertinib improved DFS. This led to the study being stopped early, leaving many unanswered questions about its potential effect on OS and its incorporation as a standard adjuvant treatment in this patient subgroup. These targeted agents are also being evaluated in locally-advanced disease, with promising results, although prospective studies with larger sample sizes are needed to confirm these results. In this article, we review the most relevant studies on the role of EGFR TKIs in the management of early-stage EGFR-mutated NSCLC.
Collapse
Affiliation(s)
- Miguel J Sotelo
- Department of Medical Oncology, Hospital María Auxiliadora; Department of Medical Oncology, Centro Oncológico Aliada; Oncological Research Unit, Clínica San Gabriel, Lima 15801, Peru
| | - José Luis García
- Department of Thoracic Surgery, Hospital Universitario La Princesa; Department of Thoracic Surgery, MD Anderson Cancer Center; Department of Thoracic Surgery, Hospital HM, Madrid 28006, Spain
| | - Cesar Torres-Mattos
- Department of Medical Oncology, Hospital Nacional Guillermo Almenara; Oncological Research Unit, Clínica San Gabriel, Lima 15033, Peru
| | - Héctor Milián
- Department of Thoracic Surgery, Hospital Universitario La Princesa, Madrid 28006, Spain
| | - Carlos Carracedo
- Department of Medical Oncology, Centro Oncológico Aliada, Lima 15036, Peru
| | | | - Xabier Mielgo-Rubio
- Department of Oncology, Hospital Universitario Fundación Alcorcón, Alcorcón 28922, Madrid, Spain
| | | | - Felipe Couñago
- Department of Radiation Oncology, Hospital Universitario Quirónsalud Madrid; Hospital La Luz; Universidad Europea de Madrid, Madrid 28223, Spain
| |
Collapse
|
28
|
Song JW, Zhu J, Wu XX, Tu T, Huang JQ, Chen GZ, Liang LY, Zhou CH, Xu X, Gong LY. GOLPH3/CKAP4 promotes metastasis and tumorigenicity by enhancing the secretion of exosomal WNT3A in non-small-cell lung cancer. Cell Death Dis 2021; 12:976. [PMID: 34671013 PMCID: PMC8528870 DOI: 10.1038/s41419-021-04265-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/30/2021] [Accepted: 10/06/2021] [Indexed: 12/21/2022]
Abstract
Cancer metastasis is the main cause of mortality associated with non-small-cell lung cancer (NSCLC), accounting for up to 70% of deaths among patients. The mechanisms underlying distal metastasis remain largely unknown. Golgi phosphoprotein 3 (GOLPH3) correlates negatively with overall survival in multiple tumors. In this study, we evaluated the function of GOLPH3 in NSCLC distal metastasis. GOLPH3 was expressed at high levels in samples from patients with NSCLC and was positively associated with clinicopathologic characteristics including clinical stage (P < 0.001), T (P = 0.001), N (P = 0.007), and M (P = 0.001) classification. Functionally, Transwell and wound-healing assays suggested that GOLPH3 overexpression enhances NSCLC cell migration and invasion abilities. Tumor-sphere formation and flow cytometry assays demonstrated that GOLPH3 overexpression enhances a stem cell-like phenotype of NSCLC cells. Metastasis models established by tail vein and intracardiac injection confirmed the pro-metastatic function of GOLPH3 in vivo. A subcutaneous tumor formation model confirmed that GOLPH3 overexpression increased the tumorigenicity of NSCLC cells. Mechanistically, gene set enrichment analysis revealed a positive association of GOLPH3 mRNA expression with WNT-activated gene signatures. Luciferase-reporter and nuclear extract assays showed that GOLPH3 overexpression enhances metastasis and tumorigenicity through activation of the WNT/β-catenin pathway. Immunoprecipitation-mass spectrometry and gene ontology analysis demonstrated that GOLPH3 interacts with cytoskeleton-associated protein 4 (CKAP4) in exosome-mediated distal metastasis. We found that GOLPH3 decreased the amount of plasma membrane-localized CKAP4 and increased the amount of exosome-localized CKAP4 to promote the formation of CKAP4-containing exosomes. Furthermore, we demonstrated that CKAP4 binds exosomal WNT3A to enhance its secretion. Therefore, the GOLPH3/CKAP4 axis plays a crucial role in promoting exosomal-WNT3A secretion to enhance and maintain the stem-like phenotype and metastasis in NSCLC, thus indicating the therapeutic potential of GOLPH3 in patients with NSCLC metastasis.
Collapse
Affiliation(s)
- Jun-Wei Song
- GuangDong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, Health Science Center, Shenzhen University, 518060, Shenzhen, Guangdong, P. R. China
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, 518060, Shenzhen, Guangdong, P. R. China
| | - Jing Zhu
- GuangDong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, Health Science Center, Shenzhen University, 518060, Shenzhen, Guangdong, P. R. China
| | - Xing-Xuan Wu
- Department of Cell Biology and Medical Genetics, School of Basic Medical Sciences, Health Science Center, Shenzhen University, 518055, Shenzhen, Guangdong, China
| | - Ting Tu
- GuangDong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, Health Science Center, Shenzhen University, 518060, Shenzhen, Guangdong, P. R. China
| | - Jing-Qiang Huang
- GuangDong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, Health Science Center, Shenzhen University, 518060, Shenzhen, Guangdong, P. R. China
| | - Guan-Zi Chen
- GuangDong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, Health Science Center, Shenzhen University, 518060, Shenzhen, Guangdong, P. R. China
| | - Li-Yin Liang
- GuangDong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, Health Science Center, Shenzhen University, 518060, Shenzhen, Guangdong, P. R. China
| | - Chun-Hui Zhou
- Guangzhou Health Science College, 510520, Guangzhou, Guangdong, P. R. China
| | - XingZhi Xu
- Department of Cell Biology and Medical Genetics, School of Basic Medical Sciences, Health Science Center, Shenzhen University, 518055, Shenzhen, Guangdong, China
- Shenzhen University-Friedrich Schiller Universität Jena Joint PhD Program in Biomedical Sciences, Health Science Center, Shenzhen University, 518055, Shenzhen, Guangdong, China
- Carson International Cancer Center, Health Science Center, Shenzhen University, 518055, Shenzhen, Guangdong, China
| | - Li-Yun Gong
- GuangDong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, Health Science Center, Shenzhen University, 518060, Shenzhen, Guangdong, P. R. China.
| |
Collapse
|
29
|
K Ca channel blockers increase effectiveness of the EGF receptor TK inhibitor erlotinib in non-small cell lung cancer cells (A549). Sci Rep 2021; 11:18330. [PMID: 34526525 PMCID: PMC8443639 DOI: 10.1038/s41598-021-97406-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 08/18/2021] [Indexed: 11/08/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) has a poor prognosis with a 5 year survival rate of only ~ 10%. Important driver mutations underlying NSCLC affect the epidermal growth factor receptor (EGFR) causing the constitutive activation of its tyrosine kinase domain. There are efficient EGFR tyrosine kinase inhibitors (TKIs), but patients develop inevitably a resistance against these drugs. On the other hand, KCa3.1 channels contribute to NSCLC progression so that elevated KCa3.1 expression is a strong predictor of poor NSCLC patient prognosis. The present study tests whether blocking KCa3.1 channels increases the sensitivity of NSCLC cells towards the EGFR TKI erlotinib and overcomes drug resistance. mRNA expression of KCa3.1 channels in erlotinib-sensitive and -resistant NSCLC cells was analysed in datasets from Gene expression omnibus (GEO) and ArrayExpress. We assessed proliferation and migration of NSCLC cells. These (live cell-imaging) experiments were complemented by patch clamp experiments and Western blot analyses. We identified three out of four datasets comparing erlotinib-sensitive and -resistant NSCLC cells which revealed an altered expression of KCa3.1 mRNA in erlotinib-resistant NSCLC cells. Therefore, we evaluated the combined effect of erlotinib and the KCa3.1 channel inhibition with sencapoc. Erlotinib elicits a dose-dependent inhibition of migration and proliferation of NSCLC cells. The simultaneous application of the KCa3.1 channel blocker senicapoc increases the sensitivity towards a low dose of erlotinib (300 nmol/L) which by itself has no effect on migration and proliferation. Partial erlotinib resistance can be overcome by KCa3.1 channel blockade. The sensitivity towards erlotinib as well as the potentiating effect of KCa3.1 blockade is further increased by mimicking hypoxia. Our results suggest that KCa3.1 channel blockade may constitute a therapeutic concept for treating NSCLC and overcome EGFR TKI resistance. We propose that this is due to complementary mechanisms of action of both blockers.
Collapse
|
30
|
Yan T, Ma G, Wang K, Liu W, Zhong W, Du J. The Immune Heterogeneity Between Pulmonary Adenocarcinoma and Squamous Cell Carcinoma: A Comprehensive Analysis Based on lncRNA Model. Front Immunol 2021; 12:547333. [PMID: 34394068 PMCID: PMC8358782 DOI: 10.3389/fimmu.2021.547333] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/13/2021] [Indexed: 01/31/2023] Open
Abstract
Adenocarcinoma (AD) and squamous cell carcinoma (SCC) are both classified as major forms of non-small cell lung cancer, but differences in clinical prognoses and molecular mechanisms are remarkable. Recent studies have supported the importance of understanding immune status in that it influences clinical outcomes of cancer, and immunotherapies based on the theory of “immune editing” have had notable clinical success. Our study aimed to identify specific long non-coding (lnc) RNAs that control key immune-related genes and to use them to construct risk models for AD and SCC. Risk scores were used to separate patients into high- and low-risk groups, and we validated the prognostic significance of both risk scores with our own cohorts. A Gene Set Enrichment Analysis suggested that the immune responses of patients in the AD high-risk group and the SCC low-risk group tended to be weakened. Evaluation of immune infiltration revealed that the degree of infiltration of dendritic cells is of particular importance in AD. In addition, prediction of responses to immune checkpoint inhibitor (ICI) treatments, based on the T Cell Immune Dysfunction and Exclusion and immunophenoscore models, indicated that deterioration of the immune microenvironment is due mainly to T cell exclusion in AD patients and T cell dysfunction in SCC patients and that high-risk patients with SCC might benefit from ICI treatment. The prediction of downstream targets via The Cancer Proteome Atlas and RNA-seq analyses of a transfected lung cancer cell line indicated that the lncRNA LINC00996 is a potential therapeutic target in AD.
Collapse
Affiliation(s)
- Tao Yan
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guoyuan Ma
- Department of Thoracic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Kai Wang
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Weidong Liu
- Department of Interventional Radiology, Shandong Medical Imaging Research Institute Affiliated to Shandong University, Jinan, China
| | - Weiqing Zhong
- Department of Radiology, The Fourth People's Hospital of Jinan, Jinan, China
| | - Jiajun Du
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Thoracic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
31
|
Pardo-Sánchez JM, Mancheño N, Cerón J, Jordá C, Ansotegui E, Juan Ó, Palanca S, Cremades A, Gandía C, Farràs R. Increased Tumor Growth Rate and Mesenchymal Properties of NSCLC-Patient-Derived Xenograft Models during Serial Transplantation. Cancers (Basel) 2021; 13:cancers13122980. [PMID: 34198671 PMCID: PMC8232339 DOI: 10.3390/cancers13122980] [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: 03/27/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) is the leading cause of cancer death worldwide. The high mortality is very often a consequence of its late diagnosis when the cancer is already locally advanced or has disseminated. Advances in the study of NSCLC tumors have been achieved by using in vivo models, such as patient-derived xenografts. Apart from drug screening, this approach may also be useful for study of the biology of the tumors. In the present study, surgically resected primary lung cancer samples (n = 33) were implanted in immunodeficient mice, and nine were engrafted successfully, including seven adenocarcinomas, one squamous-cell carcinoma, and one large-cell carcinoma. ADC tumors bearing the KRAS-G12C mutation were the most frequently engrafted in our PDX collection. Protein expression of vimentin, ezrin, and Ki67 were evaluated in NSCLC primary tumors and during serial transplantation by immunohistochemistry, using H-score. Our data indicated a more suitable environment for solid adenocarcinoma, compared to other lung tumor subtypes, to grow and preserve its architecture in mice, and a correlation between higher vimentin and ezrin expression in solid adenocarcinomas. A correlation between high vimentin expression and lung adenocarcinoma tumors bearing KRAS-G12C mutation was also observed. In addition, tumor evolution towards more proliferative and mesenchymal phenotypes was already observed in early PDX tumor passages. These PDX models provide a valuable platform for biomarker discovery and drug screening against tumor growth and EMT for lung cancer translational research.
Collapse
Affiliation(s)
- José Miguel Pardo-Sánchez
- Oncogenic Signalling Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (J.M.P.-S.); (C.G.)
| | - Nuria Mancheño
- Department of Pathology, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain;
| | - José Cerón
- Department of Thoracic Surgery, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain; (J.C.); (C.J.)
| | - Carlos Jordá
- Department of Thoracic Surgery, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain; (J.C.); (C.J.)
| | - Emilio Ansotegui
- Department of Pulmonology, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain;
| | - Óscar Juan
- Department of Medical Oncology, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain;
| | - Sarai Palanca
- Molecular Biology Unit, Service of Clinical Analysis, University and Polytechnic La Fe Hospital, 46026 Valencia, Spain;
| | - Antonio Cremades
- Department of Pathology, Hospital Universitario de la Ribera, 46600 Alzira, Spain;
| | - Carolina Gandía
- Oncogenic Signalling Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (J.M.P.-S.); (C.G.)
| | - Rosa Farràs
- Oncogenic Signalling Laboratory, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain; (J.M.P.-S.); (C.G.)
- Correspondence:
| |
Collapse
|
32
|
Pal J, Becker AC, Dhamija S, Seiler J, Abdelkarim M, Sharma Y, Behr J, Meng C, Ludwig C, Kuster B, Diederichs S. Systematic analysis of migration factors by MigExpress identifies essential cell migration control genes in non-small cell lung cancer. Mol Oncol 2021; 15:1797-1817. [PMID: 33934493 PMCID: PMC8253088 DOI: 10.1002/1878-0261.12973] [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: 01/28/2021] [Revised: 04/01/2021] [Accepted: 04/07/2021] [Indexed: 11/07/2022] Open
Abstract
Cell migration is an essential process in health and in disease, including cancer metastasis. A comprehensive inventory of migration factors is nonetheless lacking-in part due to the difficulty in assessing migration using high-throughput technologies. Hence, there are currently very few screens that systematically reveal factors controlling cell migration. Here, we introduce MigExpress as a platform for the 'identification of Migration control genes by differential Expression'. MigExpress exploits the combination of in-depth molecular profiling and the robust quantitative analysis of migration capacity in a broad panel of samples and identifies migration-associated genes by their differential expression in slow- versus fast-migrating cells. We applied MigExpress to investigate non-small cell lung cancer (NSCLC), which is the most frequent cause of cancer mortality mainly due to metastasis. In 54 NSCLC cell lines, we comprehensively determined mRNA and protein expression. Correlating the transcriptome and proteome profiles with the quantified migration properties led to the discovery and validation of FLNC, DSE, CPA4, TUBB6, and BICC1 as migration control factors in NSCLC cells, which were also negatively correlated with patient survival. Notably, FLNC was the least expressed filamin in NSCLC, but the only one controlling cell migration and correlating with patient survival and metastatic disease stage. In our study, we present MigExpress as a new method for the systematic analysis of migration factors and provide a comprehensive resource of transcriptomic and proteomic data of NSCLC cell lines related to cell migration.
Collapse
Affiliation(s)
- Jagriti Pal
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Germany
| | - Andrea C Becker
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Germany
| | - Sonam Dhamija
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Germany.,Division of RNA Biology & Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,CSIR Institute of Genomics and Integrative Biology, New Delhi, India
| | - Jeanette Seiler
- Division of RNA Biology & Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mahmoud Abdelkarim
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Germany
| | - Yogita Sharma
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Germany
| | - Jürgen Behr
- Leibniz Institute for Food Systems, Technical University of Munich, Freising, Germany.,Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technical University of Munich, Freising, Germany
| | - Chen Meng
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technical University of Munich, Freising, Germany
| | - Christina Ludwig
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technical University of Munich, Freising, Germany
| | - Bernhard Kuster
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technical University of Munich, Freising, Germany.,Chair of Proteomics and Bioanalytics, DKTK Partner Site Munich, Freising, Germany
| | - Sven Diederichs
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) - Partner Site Freiburg, Germany.,Division of RNA Biology & Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
33
|
Cao B, Wang P, Gu L, Liu J. Use of four genes in exosomes as biomarkers for the identification of lung adenocarcinoma and lung squamous cell carcinoma. Oncol Lett 2021; 21:249. [PMID: 33664813 PMCID: PMC7882885 DOI: 10.3892/ol.2021.12510] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 01/08/2021] [Indexed: 02/07/2023] Open
Abstract
The determination of biomarkers in the blood specific for lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) is crucial for the selection of effective treatment strategies and the prediction of prognosis. The purpose of the present study was to analyze the differentially expressed genes (DEGs) in LUSC and LUAD from The Cancer Genome Atlas (TCGA) database. In order to identify the potential biomarkers for non-small cell lung cancer (NSCLC) for clinical diagnosis, bioinformatics was used to analyze the DEGs of two subtypes of NSCLC, LUAD and LUSC. Exosomes were isolated from the serum of patients with LUAD or LUSC and identified using transmission electron microscopy, nanoparticle tracking analysis and western blot analysis. A total of four differential exosomal mRNAs were selected for validation with serum samples from 70 patients with NSCLC via reverse transcription-quantitative polymerase chain reaction. Receiver operating characteristic curves were established to evaluate the clinical diagnostic value of four DEGs for patients with LUAD and LUSC. The analysis based on TCGA data revealed the DEGs in LUSC and LUAD: A total of 1,619 genes were differentially expressed in patients with LUSC and LUAD. DEGs analyzed by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses revealed that inflammation-related signaling pathways, such as complement pathways, and multiple autoimmune diseases, such as systemic lupus erythematosus and asthma were mainly enriched in LUAD. The cell cycle, Hippo signaling pathway, Rap1 signaling pathway and Wnt signaling pathway were the main signaling pathways enriched in LUSC. The combination of tumor protein P63 (TP63), keratin 5 (KRT5), CEA cell adhesion molecule 6 (CEACAM6) and surfactant protein B (SFTPB) improved the specificity and sensitivity in the diagnosis of different lung cancer subtypes. Exosomal TP63, KRT5, CEACAM6 and SFTPB mRNAs can thus be used as biomarkers to differentiate between LUSC and LUAD, and may provide a novel strategy for their differential diagnosis and treatment.
Collapse
Affiliation(s)
- Bingji Cao
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Pengyu Wang
- Department of Clinical Laboratory, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Lina Gu
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Junfeng Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
34
|
Dong Y, Qiu T, Xuan Y, Liu A, Sun X, Huang Z, Su W, Du W, Yun T, Wo Y, Navarro A, Jiao W. circFBXW7 attenuates malignant progression in lung adenocarcinoma by sponging miR-942-5p. Transl Lung Cancer Res 2021; 10:1457-1473. [PMID: 33889522 PMCID: PMC8044477 DOI: 10.21037/tlcr-21-230] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background As a type of non-coding RNA, circular RNAs (circRNAs) are considered to be functional molecules associated with human cancers. An increasing number of circRNAs have been verified in malignant progression in a number of cancers. The circRNA, circFBXW7, has been proven to play an important role in tumor proliferation and metastasis. However, whether circFBXW7 influences progression in lung adenocarcinoma (LUAD) remains unclear. Methods Quantitative real-time reverse transcriptase PCR (qRT-PCR) was used to verify circFBXW7 in LUAD cell lines and LUAD tissues. Kaplan-Meier analysis was then used to compare the disease-free survival (DFS) and overall survival (OS) of these LUAD patients. The biological function of circFBXW7 was examined by overexpression and knockdown of circFBXW7 using MTT assay, EdU assay, wound-healing assay, and Transwell in vitro assays. To explore the mechanism of the circFBXW7, RNA pull-down assay, dual luciferase reporter assay, and RNA immunoprecipitation (RIP) assay were employed to examine the interaction between circFBXW7 and miR-942-5p. Western blot was used to study the fundamental proteins associated with the epithelial-mesenchymal transition (EMT) pathway. In vivo studies with BALB/c nude mice subcutaneously injected with cells stably overexpressing circFBXW7 were performed to further validate the in vitro results. Results circFBXW7 was downregulated in LUAD cell lines and tissues, and LUAD patients with lower levels had shorter DFS and OS. The in vitro study showed that circFBXW7 overexpression inhibited proliferation and migration of A549 and HCC2279 cell lines. These results were confirmed by circFBXW7 knockdown, which showed the reverse effect. The in vivo model showed that the circRNA levels influenced the tumor growth. Finally, we determined that circFBXW7 target miRNA-942-5p which regulates the EMT gene BARX2. The modulation of circFBXW7 levels produced significant changes in EMT genes in vitro and in vivo. Conclusions Our findings showed that circFBXW7 inhibits proliferation and migration by controlling the miR-942-5p/BARX2 axis in LUAD cell lines and its levels correlates with patient survival suggesting that regulating circFBXW7 could have therapeutic value in treating LUAD patients.
Collapse
Affiliation(s)
- Yanting Dong
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tong Qiu
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yunpeng Xuan
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ao Liu
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiao Sun
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhangfeng Huang
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenhao Su
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenxing Du
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tianxiang Yun
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yang Wo
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Alfons Navarro
- Molecular Oncology and Embryology Laboratory, Human Anatomy Unit, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Wenjie Jiao
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
35
|
Vinod Prabhu V, Elangovan P, Niranjali Devaraj S, Sakthivel KM. Targeting NF-κB mediated cell signaling pathway and inflammatory mediators by 1,2-diazole in A549 cells in vitro. ACTA ACUST UNITED AC 2021; 29:e00594. [PMID: 33598414 PMCID: PMC7868824 DOI: 10.1016/j.btre.2021.e00594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/15/2021] [Accepted: 01/25/2021] [Indexed: 11/30/2022]
Abstract
1,2-Diazole suppresses TNF-α induced MMP-2 expression. 1,2-Diazole abrogate NF-κB activation and regulate cytokines. It exhibits potent in vitro anticancer effect against A549 cells.
Lung cancer is the leading cause of cancer deaths globally. The objective of this study was to investigate the effect of 1,2-diazole (pyrazole) as an anti-cancer drug on human non-small cell lung carcinoma A549 cells. We attempt to examine the expression level of pro-inflammatory proteins such as TNF-α, NF-κB-p65, MMP-2 and E-Cadherin which are commonly associated with an inflammatory response in epithelial cells and apoptosis in A549 cells. The LPS-induced cytokines and inflammatory mediators include TNF-α, IL-6, iNOS and COX-2 levels in A549 cells and the effect of pyrazole was studied. The present study reveals that, pyrazole inhibits A549 cells by suppressing TNF-α induced MMP-2 expression, thereby inhibiting the nuclear translocation of NF-κB-p65. Pyrazole significantly up-regulate the E-cadherin level and down-regulated MMP-2 expression that could probably preventing A549 cancer cells to invade. The study further substantiated the anti-cancer property of pyrazole by regulating the above mentioned level of LPS-induced cytokines and inflammatory mediators. The observations of the present study open a possibility for the development of an effective therapeutic agent that targets inflammatory and signaling pathway mediators to challenge human non-small cell lung carcinoma.
Collapse
Affiliation(s)
- Venugopal Vinod Prabhu
- Department of Biochemistry, University of Madras, Guindy Campus, Chennai, 600025, Tamil Nadu, India
- Corresponding author.
| | - Perumal Elangovan
- Department of Biochemistry, University of Madras, Guindy Campus, Chennai, 600025, Tamil Nadu, India
| | | | | |
Collapse
|
36
|
Chen X, Wang Y, Qu X, Bie F, Wang Y, Du J. TRIM58 is a prognostic biomarker remodeling the tumor microenvironment in KRAS-driven lung adenocarcinoma. Future Oncol 2021; 17:565-579. [PMID: 33406903 DOI: 10.2217/fon-2020-0645] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: To comprehensively analyze the expression profiles of ubiquitin-related genes (URGs) and determine potential biomarkers in KRAS-driven lung adenocarcinoma (LUAD). Materials & methods: Differential expression analyses were performed between KRAS-wild and KRAS-mutant LUAD samples from The Cancer Genome Atlas database, and 34 URGs were screened out. ESTIMATE and CIBERSORT methods were used to calculate the ratio of immune and stromal components. Results & conclusion: TRIM58 was positively correlated with abundances of M2 macrophages and resting mast cells and negatively correlated with follicular helper T-cell abundances in KRAS-driven LUAD. TRIM58 was a potential prognosis-associated indicator for tumor microenvironment modulation and played a key role in TME-specific AS landscapes alterations in KRAS-driven LUAD.
Collapse
Affiliation(s)
- Xiaowei Chen
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Yu Wang
- Department of Respiratory Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, China
| | - Xiao Qu
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Fenglong Bie
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Yadong Wang
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| | - Jiajun Du
- Institute of Oncology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.,Department of Thoracic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China
| |
Collapse
|
37
|
Nishio M, Barlesi F, West H, Ball S, Bordoni R, Cobo M, Longeras PD, Goldschmidt J, Novello S, Orlandi F, Sanborn RE, Szalai Z, Ursol G, Mendus D, Wang L, Wen X, McCleland M, Hoang T, Phan S, Socinski MA. Atezolizumab Plus Chemotherapy for First-Line Treatment of Nonsquamous NSCLC: Results From the Randomized Phase 3 IMpower132 Trial. J Thorac Oncol 2020; 16:653-664. [PMID: 33333328 DOI: 10.1016/j.jtho.2020.11.025] [Citation(s) in RCA: 266] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/16/2020] [Accepted: 11/26/2020] [Indexed: 01/04/2023]
Abstract
INTRODUCTION We report the final results of the phase 3 IMpower132 study evaluating atezolizumab plus carboplatin or cisplatin plus pemetrexed (APP) in patients with nonsquamous NSCLC. METHODS Chemotherapy-naive patients with stage IV nonsquamous NSCLC without sensitizing EGFR or ALK genetic alterations were randomized in a one-to-one ratio to receive four or six cycles of carboplatin or cisplatin plus pemetrexed (PP) or APP every 3 weeks, followed by maintenance therapy with atezolizumab plus pemetrexed or pemetrexed alone. Co-primary end points were overall survival (OS) and investigator-assessed progression-free survival (PFS). RESULTS The intention-to-treat population included 578 patients (APP, n = 292; PP, n = 286). At the primary PFS analysis (May 22, 2018; median follow-up, 14.8 mo), APP exhibited significant PFS improvement versus PP (median = 7.6 versus 5.2 mo, stratified hazard ratio [HR] = 0.60, 95% confidence interval [CI]: 0.49-0.72, p < 0.0001). OS for the APP group was numerically better but not statistically significant at the interim (May 22, 2018; median = 18.1 versus 13.6 mo, stratified HR = 0.81, 95% CI: 0.64-1.03, p = 0.0797) and final analyses (July 18, 2019; median = 17.5 versus 13.6 mo; stratified HR = 0.86, 95% CI: 0.71-1.06, p = 0.1546). The OS and PFS results favored APP versus PP across subgroups. Grade 3 or 4 treatment-related adverse events occurred in 54.6% (APP) and 40.1% (PP) of patients; grade 5 treatment-related events occurred in 3.8% and 2.9%, respectively. CONCLUSIONS IMpower132 met its co-primary PFS end point but not its co-primary OS end point, with numerical improvement for OS in the APP arm. APP had a manageable safety profile, with no new or unexpected safety signals identified.
Collapse
Affiliation(s)
- Makoto Nishio
- The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Fabrice Barlesi
- Department of Medical Oncology, Cancer Research Center of Marseille, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Assistance Publique-Hopitaux de Marseille, Campus Timone, Centres Hospitaliers et Universitaires Nord, Aix Marseille University, Marseille, France; Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Howard West
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Simon Ball
- Department of Oncology, Queen's Hospital, Rom Valley Way, Romford, United Kingdom
| | | | - Manuel Cobo
- Unidad de Gestión Clínica Intercentros de Oncología Médica. Hospitales Universitarios Regional y Virgen de la Victoria, Málaga, Spain
| | | | | | - Silvia Novello
- Department of Oncology, University of Turin, Orbassano, Italy
| | - Francisco Orlandi
- Department of Medical Oncology, Instituto Nacional del Tόrax, Santiago, Chile
| | - Rachel E Sanborn
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon
| | - Zsuzsanna Szalai
- Department of Pulmonology, Petz Aladár County Teaching Hospital, Vasvári Pál, Hungary
| | | | - Diana Mendus
- Genentech, Inc., South San Francisco, California
| | - Lijia Wang
- Genentech, Inc., South San Francisco, California
| | - Xiaohui Wen
- Genentech, Inc., South San Francisco, California
| | | | - Tien Hoang
- Genentech, Inc., South San Francisco, California
| | - See Phan
- Genentech, Inc., South San Francisco, California
| | - Mark A Socinski
- Department of Medical Oncology, AdventHealth Cancer Institute, Orlando, Florida
| |
Collapse
|
38
|
Wang J, Chen H, Li W, Shan L. Cordyceps acid alleviates lung cancer in nude mice. J Biochem Mol Toxicol 2020; 35:e22670. [PMID: 33251677 DOI: 10.1002/jbt.22670] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/26/2020] [Accepted: 11/04/2020] [Indexed: 12/30/2022]
Abstract
The aim of this study is to examine the effect of cordyceps acid (CA) on lung cancer in mice. A lung cancer animal model was established by inoculating lung cancer cells under the armpit of nude mice. The mental state, body weight, and tumor growth of nude mice were recorded in detail. The levels of Nrf-2/HO-1/NLRP3/NF-κB pathway and apoptosis in tumor tissues of nude mice were detected by the Western blot analysis and immunohistochemical methods. Our results show that CA inhibited lung cancer by regulating the Nrf-2/HO-1/NLRP3/NF-κB signal. In summary, CA has an obvious tumor inhibiting effect on lung cancer via regulation of the Nrf-2/HO-1/NLRP3/NF-κB signal.
Collapse
Affiliation(s)
- Jing Wang
- School of Biology and Food Engineering, Institute of Pharmaceutical Biotechnology, Suzhou University, Suzhou, China
| | - Hongling Chen
- School of Biology and Food Engineering, Institute of Pharmaceutical Biotechnology, Suzhou University, Suzhou, China
| | - Wanrong Li
- School of Biology and Food Engineering, Institute of Pharmaceutical Biotechnology, Suzhou University, Suzhou, China
| | - Lingling Shan
- School of Biology and Food Engineering, Institute of Pharmaceutical Biotechnology, Suzhou University, Suzhou, China
| |
Collapse
|
39
|
Qian H, Deng J, Lu C, Hou G, Zhang H, Zhang M, Fang Z, Lv XD. Ceramide synthases: insights into the expression and prognosis of lung cancer. Exp Lung Res 2020; 47:37-53. [PMID: 33183094 DOI: 10.1080/01902148.2020.1844345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
CerSs (ceramide synthases), a group of enzymes that catalyze the formation of ceramides from sphingoid base and acyl-CoA substrates. As far, six types of CerSs (CerS1-CerS6) have been found in mammals. Each of these enzymes have unique characteristics, but maybe more noteworthy is the ability of individual CerS isoform to produce a ceramide with a characteristic acyl chain distribution. As key regulators of sphingolipid metabolism, CerSs highlight their unique characteristics and have emerging roles in regulating programmed cell death, cancer and many other aspects of biology. However, the role of CerSs in lung cancer has not been fully elucidated. In this study, there was no significant change in the sequence or copy number of CerSs gene, which could explain the stability of malignant tumor development through COSMIC database. In addition, gene expression in lung cancer was examined using the OncomineTM database, and the prognostic value of each gene in non-small cell lung cancer (NSCLC) was analyzed by Kaplan-Meier analysis. The results showed that high mRNA expression levels of CerS2, CerS3, CerS4 and CerS5 in all NSCLC patients were associated with improved prognosis. Among them, CerS2 and CerS5 are also highly expressed in adenocarcinoma (Ade), but not in squamous cell carcinoma (SCC). In contrast, high or low expression of CerS1 and CerS6 no difference was observed in patients with NSCLC, Ade and SCC. Integrated the data of this study suggested that these CerSs may be a potential tumor markers or drug target of new research direction.
Collapse
Affiliation(s)
- Huijiang Qian
- Department of Respiratory, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University
| | - Jingjing Deng
- Department of Respiratory, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University
| | - Chao Lu
- Department of Cardiothoracic Surgery, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University
| | - Gouxin Hou
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing, Zhejiang, P.R. China
| | - Hualiang Zhang
- Department of Respiratory, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University
| | - Ming Zhang
- Department of Respiratory, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University
| | - Zhixian Fang
- Department of Respiratory, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University
| | - Xiao-Dong Lv
- Department of Respiratory, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University
| |
Collapse
|
40
|
Li R, Liu B, Liu Y, Liu Y, He Y, Wang D, Sun Y, Xu Y, Yu Q. Elevated serum lipid level can serve as early signal for metastasis for Non-Small Cell Lung Cancer patients: A retrospective nested case-control study. J Cancer 2020; 11:7023-7031. [PMID: 33123292 PMCID: PMC7592011 DOI: 10.7150/jca.48322] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/24/2020] [Indexed: 12/24/2022] Open
Abstract
Objective: To investigate the association between serum lipid levels in patients with primary non-small cell lung cancer and the risk of developing metastases, a retrospective cohort-based nested case-control study was conducted. Material and method: Patients with primary non-small cell lung cancer admitted to the First and the Third Hospitals of Jilin University from January 2008 through December 2015 were recruited retrospectively based on their electronic medical records. A total of 524 patients were initially considered, consisting of 138 in the case group and 386 as control. Out of these, 110 were finally included in the case group and 110 as control based on additional selection criteria. The following information is collected from all the patients, total cholesterol (TC), low-density lipoprotein (LDL-C), high density lipoprotein (HDL-C) and triglyceride (TG). Logistic regressions were conducted to estimate the odds ratios (ORs) and 95% confidence intervals (95% CI) for non-small cell lung cancer (NSCLC) patients to have metastasis risk when having elevated serum lipid levels. Restricted cubic spline (RCS) curves were used to demonstrate the association between serum lipid levels and the risk of metastasis. Results: Patients with high TC level (P = 0.025, 0R = 1.35, 95% CI: 1.03-1.74) and patients with high LDL-C level (Q4: > 3.47 vs Q1: ≤2.54, P = 0.002, OR = 3.92, 95% CI: 1.31-11.77) are found to have an increased metastasis risk; and their dose-response relationship was validated by our restricted cubic spline analysis (TC: P overall association=0.02, P non-linear association = 0.73; LDL-C: P overall association=0.02, P non-linear association = 0.10). These associations were statistically significant, particularly in men who smoked, never drank, and were 65 years of age or younger. In addition, patients with simultaneously high levels of TC and LDL-C have a 60% increased risk of metastasis compared with patients with high levels of TC and normal LDL-C. Conclusion: Dyslipidemia may be a risk factor for metastasis among NSCLC patients. Examination of serum lipid level on a regular basis can provide early signal of metastasis for NSCLC patients.
Collapse
Affiliation(s)
- Rixin Li
- Cancer System Biology Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Bin Liu
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yumei Liu
- Cancer System Biology Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yang Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Yang He
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Duo Wang
- Department of Hand and Foot Surgery, The First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yunxiang Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Ying Xu
- Cancer System Biology Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
| | - Qiong Yu
- Cancer System Biology Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, China
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
41
|
Zeng Z, Yang F, Wang Y, Zhao H, Wei F, Zhang P, Zhang X, Ren X. Significantly different immunological score in lung adenocarcinoma and squamous cell carcinoma and a proposal for a new immune staging system. Oncoimmunology 2020; 9:1828538. [PMID: 33101777 PMCID: PMC7553570 DOI: 10.1080/2162402x.2020.1828538] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
TNM stage is not enough to accurately predict the prognosis of patients with non-small cell lung cancer (NSCLC). This study aimed to establish the immunological score (IS) in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), separately, and propose a new staging system in NSCLC. We used the multiplex fluorescent immunohistochemistry (mIHC) technology to detect 17 immune biomarkers of 304 patients with NSCLC. The LASSO-COX regression model was used to establish the ISNSCLC in the training cohorts. The ISNSCLC was then validated in the validation cohort. The constructed ISLUAD contained three immune features: CD4+CD73+ core of tumor (CT), PD-L1+ CT, and IDO+ invasive margin (IM). ISLUSC also contained two immune features: CD8+CD39-CD73- CT, CD8+Tim-3+ IM. In the training cohort, significant prognostic differences were found upon comparing low-ISNSCLC patients with high-ISNSCLC patients. For LUAD, the 5-y disease-free survival (DFS) rates were 54.7% vs. 8.1% and the 5-y overall survival (OS) rates were 82.4% vs. 36% (all P< .0001). For LUSC, the 5-y DFS rates were 74.0% vs. 14.7% and the 5-y OS rates were 78.2% vs. 17.6% (all P< .0001). Multivariate analyses indicated that ISNSCLC was an independent indicator for prognosis. Finally, we combined ISNSCLC with clinicopathological factors to establish a TN-I staging system and two nomogram models for clinical use. The TN-I stage had better prediction accuracy than TNM stage. The newly established ISLUAD and ISLUSC were completely different, and both were excellent indicators for the prognostic prediction. The TN-I stage could effectively improve prognostic accuracy and facilitate clinical application. Abbreviations NSCLC, non-small cell lung cancer; IS, immunological score; mIHC, multiplex fluorescent immunohistochemistry; LUAD, lung adenocarcinoma; LUSC, lung squamous cell carcinoma; CT, core of tumor; IM, invasive margin; DFS, disease-free survival; OS, overall survival; SITC, the Society for Immunotherapy of Cancer; FFPE, formalin-fixed paraffin-embedded; MWT, microwave treatment; DCA, decision curve analysis; ROC, receiver operating characteristic; AUC, area under the curve; EGFR, epidermal growth factor receptor.
Collapse
Affiliation(s)
- Ziqing Zeng
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China,National Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China,Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Fan Yang
- National Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China,Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China,Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yunliang Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China,National Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China,Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China,Department of Oncology, First Central Hospital of Baoding of Hebei Province, Baoding, China
| | - Hua Zhao
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China,National Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China,Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Feng Wei
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China,National Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China,Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Peng Zhang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China,National Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China,Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xiying Zhang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China,National Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China,Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China,National Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China,Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China,Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China,CONTACT Xiubao Ren Department of Biotherapy and Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin300060, China
| |
Collapse
|
42
|
Li J, Guo Q, Lei X, Zhang L, Su C, Liu Y, Zhou W, Chen H, Wang H, Wang F, Yan Y, Zhang J. Pristimerin induces apoptosis and inhibits proliferation, migration in H1299 Lung Cancer Cells. J Cancer 2020; 11:6348-6355. [PMID: 33033518 PMCID: PMC7532514 DOI: 10.7150/jca.44431] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 07/26/2020] [Indexed: 12/16/2022] Open
Abstract
Background: The natural occurring pristimerin, a quinonemethide triterpenoid, is extracted from a variety of species of the Celastraceae and Hippocrateaceae family. This research investigated the in vitro anti-cancer potential of pristimerin on NSCLC cells NCI-H1299 and elucidated the molecular mechanism. Methods: Cell growth inhibition by pristimerin was assessed using the MTT assay. Apoptosis was detected using the Annexin V/propidium iodide (PI) test. The colony forming assay was used to investigate the anti-proliferative effects of pristimerin. Wound healing assay and the transwell cell migration assay were utilized to determine the inhibitory effects of migration and invasion, respectively. Western blot was used to detect the protein expression, and real-time-quantitative (RT-q) PCR was used to analyze the mRNA expression. Results: The results showed that pristimerin inhibited the proliferation of H1299 cells with an IC50 value of 2.2 ± 0.34 µM and induced apoptosis in a dose-dependent manner. The colony formation ability was reduced in a dose-dependent manner. A marked inhibition of migration and invasion against H1299 cells was observed in a dose- or time-dependent manner. Moreover, the decreased protein levels of vimentin, F-actin, integrin β1, matrix metalloproteinase (MMP2) and Snail revealed the potential inhibition of epithelial-to-mesenchymal transition (EMT). The regulated mRNA levels of integrin β1, MMP2 and Snail indicated the great potential in the treatment of NSCLC. Conclusion: In conclusion, our study demonstrated that pristimerin suppressed NSCLC cells NCI-H1299 in vitro, exhibited potent activities of proliferation inhibition and apoptosis induction. Furthermore, the treatment of pristimerin decreased migration and invasion of H1299, which was correlated with EMT-related proteins and mRNA.
Collapse
Affiliation(s)
- Jiajun Li
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Qiaoru Guo
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Xueping Lei
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Lingling Zhang
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Chaoyue Su
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Yun Liu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Wenmin Zhou
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| | - Hubiao Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, P. R. China
| | - Hui Wang
- Guangzhou Institute of Pediatrics/Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, P. R. China
| | - Fenghua Wang
- Guangzhou Institute of Pediatrics/Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, P. R. China
| | - Yanyan Yan
- Institute of Immunology and School of Medicine, Shanxi Datong University, Datong 037009, P. R. China
| | - Jianye Zhang
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P. R. China
| |
Collapse
|
43
|
Han Y, Ma Y, Wu Z, Zhang F, Zheng D, Liu X, Tao L, Liang Z, Yang Z, Li X, Huang J, Guo X. Histologic subtype classification of non-small cell lung cancer using PET/CT images. Eur J Nucl Med Mol Imaging 2020; 48:350-360. [PMID: 32776232 DOI: 10.1007/s00259-020-04771-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/10/2020] [Indexed: 12/20/2022]
Abstract
PURPOSES To evaluate the capability of PET/CT images for differentiating the histologic subtypes of non-small cell lung cancer (NSCLC) and to identify the optimal model from radiomics-based machine learning/deep learning algorithms. METHODS In this study, 867 patients with adenocarcinoma (ADC) and 552 patients with squamous cell carcinoma (SCC) were retrospectively analysed. A stratified random sample of 283 patients (20%) was used as the testing set (173 ADC and 110 SCC); the remaining data were used as the training set. A total of 688 features were extracted from each outlined tumour region. Ten feature selection techniques, ten machine learning (ML) models and the VGG16 deep learning (DL) algorithm were evaluated to construct an optimal classification model for the differential diagnosis of ADC and SCC. Tenfold cross-validation and grid search technique were employed to evaluate and optimize the model hyperparameters on the training dataset. The area under the receiver operating characteristic curve (AUROC), accuracy, precision, sensitivity and specificity was used to evaluate the performance of the models on the test dataset. RESULTS Fifty top-ranked subset features were selected by each feature selection technique for classification. The linear discriminant analysis (LDA) (AUROC, 0.863; accuracy, 0.794) and support vector machine (SVM) (AUROC, 0.863; accuracy, 0.792) classifiers, both of which coupled with the ℓ2,1NR feature selection method, achieved optimal performance. The random forest (RF) classifier (AUROC, 0.824; accuracy, 0.775) and ℓ2,1NR feature selection method (AUROC, 0.815; accuracy, 0.764) showed excellent average performance among the classifiers and feature selection methods employed in our study, respectively. Furthermore, the VGG16 DL algorithm (AUROC, 0.903; accuracy, 0.841) outperformed all conventional machine learning methods in combination with radiomics. CONCLUSION Employing radiomic machine learning/deep learning algorithms could help radiologists to differentiate the histologic subtypes of NSCLC via PET/CT images.
Collapse
Affiliation(s)
- Yong Han
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Yuan Ma
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Zhiyuan Wu
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Feng Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Deqiang Zheng
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Xiangtong Liu
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Lixin Tao
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China.,Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Zhigang Liang
- Department of Nuclear Medicine, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research, Department of Nuclear Medicine, Peking University Cancer Hospital, Beijing, China
| | - Xia Li
- Department of Mathematics and Statistics, La Trobe University, Melbourne, Victoria, Australia
| | - Jian Huang
- School of Mathematical Sciences, University College Cork, Cork, Ireland
| | - Xiuhua Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China. .,Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China.
| |
Collapse
|
44
|
Park HJ, Park SH. Induction of cytoprotective autophagy by morusin via AMP-activated protein kinase activation in human non-small cell lung cancer cells. Nutr Res Pract 2020; 14:478-489. [PMID: 33029288 PMCID: PMC7520565 DOI: 10.4162/nrp.2020.14.5.478] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 03/30/2020] [Accepted: 06/04/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND/OBJECTIVES Morusin, a marker component of Morus alba L., possesses anti-cancer activity. The objective of this study was to determine autophagy-inducing effect of morusin in non-small cell lung cancer (NSCLC) cells and investigate the underlying mechanism. SUBJECTS/METHODS Autophagy induction and the expression of autophagy-related proteins were analyzed by LC3 immunofluorescence and western blot, respectively. The role of autophagy and AMP-activated protein kinase (AMPK) was determined by treating NSCLC cells with bafilomycin A1, an autophagy inhibitor, and compound C, an AMPK inhibitor. Cytotoxicity and apoptosis induction were determined by MTT assay, trypan blue exclusion assay, annexin V-propidium iodide (PI) double staining assay, and cell cycle analysis. RESULTS Morusin increased the formation of LC3 puncta in the cytoplasm and upregulated the expression of autophagy-related 5 (Atg5), Atg12, beclin-1, and LC3II in NSCLC cells, demonstrating that morusin could induce autophagy. Treatment with bafilomycin A1 markedly reduced cell viability but increased proportions of sub-G1 phase cells and annexin V-positive cells in H460 cells. These results indicate that morusin can trigger autophagy in NSCLC cells as a defense mechanism against morusin-induced apoptosis. Furthermore, we found that AMPK and its downstream acetyl-CoA carboxylase (ACC) were phosphorylated, while mammalian target of rapamycin (mTOR) and its downstream p70S6 kinase (p70S6K) were dephosphorylated by morusin. Morusin-induced apoptosis was significantly increased by treatment with compound C in H460 cells. These results suggest that morusin-induced AMPK activation could protect NSCLC cells from apoptosis probably by inducing autophagy. CONCLUSIONS Our findings suggest that combination treatment with morusin and autophagy inhibitor or AMPK inhibitor might enhance the clinical efficacy of morusin for NSCLC.
Collapse
Affiliation(s)
- Hyun-Ji Park
- Department of Pathology, College of Korean Medicine, Dong-Eui University, Busan 47227, Korea
| | - Shin-Hyung Park
- Department of Pathology, College of Korean Medicine, Dong-Eui University, Busan 47227, Korea
| |
Collapse
|
45
|
Cui Y, Li F, Xie Q, Zhao S, Guo T, Guo P, Hu S, Hao J, Tian C, Yu W, Li Z, Fang L, Zhao L, Chen M, Wu T, Gu C. YBX1 mediates autophagy by targeting p110β and decreasing the sensitivity to cisplatin in NSCLC. Cell Death Dis 2020; 11:476. [PMID: 32561752 PMCID: PMC7305216 DOI: 10.1038/s41419-020-2555-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/21/2020] [Accepted: 04/22/2020] [Indexed: 01/26/2023]
Abstract
Y-box binding protein 1 (YBX1) is involved in the development of multiple types of tumors. However, the relationship between YBX1 and autophagy in non-small cell lung cancer (NSCLC) remains unclear. In this study, we analyzed the expression and clinical significance of YBX1 and markers of autophagy (LC3I/II) in NSCLC and examined their roles in regulating sensitivity to cisplatin in NSCLC. The retrospective analysis of patients with NSCLC indicated that YBX1 was positively correlated with autophagy. Increased levels of YBX1 or autophagy also observed in NSCLC cells compared with those in 16HBE cells. Compared to the controls, the knockdown of YBX1 expression suppressed autophagy, increased drug sensitivity and promoted apoptosis in response to cisplatin in NSCLC cells by targeting the p110β promoter and inhibiting p110β/Vps34/beclin1 signaling pathways. We also demonstrated in an in vivo study that the overexpressed YBX1 effectively increased NSCLC growth and progression and decreased the sensitivity to cisplatin by inducing autophagy in a xenograft tumor model, and these effects were concomitant with the increasing of p110β and beclin1 expression. Collectively, these results show that YBX1 plays an essential role in autophagy in NSCLC.
Collapse
Affiliation(s)
- Yanwei Cui
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
- Departments of Respiratory Medicine, Zhongshan Hospital, Dalian Univerdity, 116011, Dalian, China
| | - Fengzhou Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Qiang Xie
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Shilei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Tao Guo
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Ping Guo
- Institute of Cancer Stem Cell, Dalian Medical University, 116011, Dalian, China
| | - Sheng Hu
- Institute of Cancer Stem Cell, Dalian Medical University, 116011, Dalian, China
| | - Jiaojiao Hao
- Institute of Cancer Stem Cell, Dalian Medical University, 116011, Dalian, China
| | - Chunfang Tian
- Institute of Cancer Stem Cell, Dalian Medical University, 116011, Dalian, China
| | - Wendan Yu
- Institute of Cancer Stem Cell, Dalian Medical University, 116011, Dalian, China
| | - Zhuoshi Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Lei Fang
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Lei Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China
| | - Manyu Chen
- Institute of Cancer Stem Cell, Dalian Medical University, 116011, Dalian, China
| | - Taihua Wu
- Departments of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China.
| | - Chundong Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Dalian Medical University, 116011, Dalian, China.
| |
Collapse
|
46
|
Brunetti G, Belisario DC, Bortolotti S, Storlino G, Colaianni G, Faienza MF, Sanesi L, Alliod V, Buffoni L, Centini E, Voena C, Pulito R, Novello S, Ingravallo G, Rizzi R, Mori G, Reseland JE, Ware CF, Colucci S, Ferracini R, Grano M, Roato I. LIGHT/TNFSF14 Promotes Osteolytic Bone Metastases in Non-small Cell Lung Cancer Patients. J Bone Miner Res 2020; 35:671-680. [PMID: 31826304 DOI: 10.1002/jbmr.3942] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 12/03/2019] [Accepted: 12/08/2019] [Indexed: 12/24/2022]
Abstract
Tumor necrosis factor superfamily member 14 (TNFSF14), LIGHT, is a component of the cytokine network that regulates innate and adaptive immune responses, which promote homeostasis of lymphoid organs, liver, and bone. Metastatic tumors often disrupt the tissue microenvironment, thus altering the homeostasis of the invaded organ; however, the underlying mechanisms required further studies. We investigated the role of LIGHT in osteolytic bone disease induced by metastatic non-small cell lung cancer (NSCLC). Patients diagnosed with NSCLC bone metastasis show significantly higher levels of LIGHT expressed in monocytes compared with non-bone metastatic tumors and healthy controls. Serum LIGHT levels were also higher in patients with bone metastases than in controls, suggesting a role for LIGHT in stimulating osteoclast precursors. In bone metastatic patients, we also detected increased RNA expression and serum RANKL levels, thus by adding anti-LIGHT or RANK-fragment crystallizable region (RANK-Fc) in PBMC cultures, a significant inhibition of osteoclastogenesis was observed. To model this observation in mice, we used the mouse lung cancer cell line LLC-1. After intratibial implantation, wild-type mice showed an increased number of osteoclasts but reduced numbers of osteoblasts and decreased osteoid formation. In contrast, Tnfsf14-/- mice showed no significant bone loss or other changes in bone homeostasis associated with this model. These data indicate LIGHT is a key control mechanism for regulating bone homeostasis during metastatic invasion. Thus, LIGHT may be a novel therapeutic target in osteolytic bone metastases. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Giacomina Brunetti
- Department of Basic and Medical Sciences, Neurosciences and Sense Organs, Section of Human Anatomy and Histology, University of Bari, Bari, Italy
| | - Dimas C Belisario
- Center for Experimental Research and Medical Studies (CeRMS), A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
| | - Sara Bortolotti
- Department of Emergency and Organ Transplantation, Section of Human Anatomy and Histology, University of Bari, Bari, Italy
| | - Giuseppina Storlino
- Department of Emergency and Organ Transplantation, Section of Human Anatomy and Histology, University of Bari, Bari, Italy
| | - Graziana Colaianni
- Department of Emergency and Organ Transplantation, Section of Human Anatomy and Histology, University of Bari, Bari, Italy
| | - Maria F Faienza
- Department of Biomedical Science and Human Oncology, University of Bari, Bari, Italy
| | - Lorenzo Sanesi
- Department of Emergency and Organ Transplantation, Section of Human Anatomy and Histology, University of Bari, Bari, Italy
| | - Valentina Alliod
- Department of Oncological Sciences, University of Turin Medical School, Turin, Italy
| | - Lucio Buffoni
- Department of Oncological Sciences, University of Turin Medical School, Turin, Italy
| | - Elisa Centini
- Center for Experimental Research and Medical Studies (CeRMS), A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
| | - Claudia Voena
- Center for Experimental Research and Medical Studies (CeRMS), A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Roberta Pulito
- Center for Experimental Research and Medical Studies (CeRMS), A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
| | - Silvia Novello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Giuseppe Ingravallo
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Rita Rizzi
- Department of Emergency and Organ Transplantation, University of Bari, Bari, Italy
| | - Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Janne E Reseland
- Department of Biomaterials, Institute for Clinical Dentistry, University of Oslo Blindern, Oslo, Norway
| | - Carl F Ware
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Silvia Colucci
- Department of Basic and Medical Sciences, Neurosciences and Sense Organs, Section of Human Anatomy and Histology, University of Bari, Bari, Italy
| | - Riccardo Ferracini
- Department of Surgical Sciences (DISC), Orthopaedic Clinic-IRCCS, A.O.U. San Martino, Genoa, Italy
| | - Maria Grano
- Department of Emergency and Organ Transplantation, Section of Human Anatomy and Histology, University of Bari, Bari, Italy
| | - Ilaria Roato
- Center for Experimental Research and Medical Studies (CeRMS), A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy
| |
Collapse
|
47
|
Shen DJ, Jiang YH, Li JQ, Xu LW, Tao KY. The RNA-binding protein RBM47 inhibits non-small cell lung carcinoma metastasis through modulation of AXIN1 mRNA stability and Wnt/β-catentin signaling. Surg Oncol 2020; 34:31-39. [PMID: 32891348 DOI: 10.1016/j.suronc.2020.02.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/29/2020] [Accepted: 02/14/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Non-small-cell lung cancer (NSCLC) remains a highly prevalent and deadly form of cancer, with efforts to better understand the molecular basis of the progression of this disease being essential to its effective treatment. Several recent studies have highlighted the ability of RNA-binding proteins (RBPs) to regulate a wide range of cellular processes in both healthy and pathogenic contexts. Among these RBPs, RNA binding motif protein 47 (RBM47) has recently been identified as a tumor suppressor in both breast and colon cancers, whereas its role in NSCLC is poorly understood. METHODS RBM47 expression in NSCLC samples was evaluated by RT-PCR, western blotting and immunohistochemistry analysis. Molecular and cellular techniques including lentiviral vector-mediated knockdown were used to elucidate the functions and mechanisms of RBM47. RESULTS This study sought to analyze the expression and role of RBM47 in NSCLC. In the present study, we observed reduced levels of RBM47 expression in NSCLC, with these reductions corresponding to a poorer prognosis and more advanced disease including a higher TNM stage (p = 0.022), a higher likelihood of tumor thrombus (p = 0.001), and pleural invasion (p = 0.033). Through functional analyses in vitro and in vivo, we further demonstrated that these RBP was able to disrupt the proliferation, migration, and invasion of NSCLC cells. At a molecular level, we determined that RBM47 was able to bind the AXIN1 mRNA, stabilizing it and thereby enhancing the consequent suppression of Wnt/β-catentin signaling. CONCLUSION Together our findings reveal that RBM47 targets AXIN1 in order to disrupt Wnt/β-catenin signaling in NSCLC and thereby disrupting tumor progression. These results thus offer new insights into the molecular biology of NSCLC, and suggest that RBM47 may also have value as a prognostic biomarker and/or therapeutic target in NSCLC patients.
Collapse
Affiliation(s)
- Di-Jian Shen
- Department of Thoracic Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Gongshu District, Hangzhou, 310022, China
| | - You-Hua Jiang
- Department of Thoracic Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Gongshu District, Hangzhou, 310022, China
| | - Jian-Qiang Li
- Department of Thoracic Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Gongshu District, Hangzhou, 310022, China
| | - Li-Wei Xu
- Department of Thoracic Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Gongshu District, Hangzhou, 310022, China
| | - Kai-Yi Tao
- Department of Thoracic Surgery, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, No. 1 Banshan East Road, Gongshu District, Hangzhou, 310022, China.
| |
Collapse
|
48
|
Han Z, Li Y, Zhang J, Guo C, Li Q, Zhang X, Lan Y, Gu W, Xing Z, Liang L, Li M, Mi S. Tumor-derived circulating exosomal miR-342-5p and miR-574-5p as promising diagnostic biomarkers for early-stage Lung Adenocarcinoma. Int J Med Sci 2020; 17:1428-1438. [PMID: 32624699 PMCID: PMC7330662 DOI: 10.7150/ijms.43500] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/15/2020] [Indexed: 01/08/2023] Open
Abstract
Lung cancer has been the leading cause of cancer morbidity and mortality in recent years. Most lung cancers are often asymptomatic until advanced or metastatic stage. Therefore, looking for the diagnostic biomarker for early-stage lung cancer is quite significant. Circulating exosomal microRNAs (miRNAs) have been reported to be the diagnostic and prognostic markers of various cancers. Here, we obtained circulating exosomal miRNA repertoires of 7 early-stage lung adenocarcinoma patients including pre-operation and post-operation (LA-pre and LA-post) and 7 heathy controls (HCs) by next generation sequence (NGS) and selected miR-342-5p, miR-574-5p and miR-222-3p to validate in ampliative samples by reverse transcription-quantitative PCR (RT-qPCR). Circulating exosomal miR-342-5p, miR-574-5p and miR-222-3p not only significantly elevated in LA patients (n = 56) compared with HCs (n = 40), but also significantly decreased after tumor resection when analyzed 51 paired pre- and post-operation samples. Furthermore, miR-342-5p and miR-574-5p, but not miR-222-3p, had a significantly elevated expression level in carcinoma tissue compared with adjacent non-cancerous tissue (n = 8). The receiver operating characteristic (ROC) curve showed the area under the curve (AUC) of combined miR-342-5p and miR-574-5p was 0.813 (95% CI: 0.7249 to 0.9009) with sensitivity and specificity of 80.0% and 73.2% respectively. In summary, circulating exosomal miR-342-5p and miR-574-5p have potential to serve as novel diagnostic biomarkers for early-stage LA.
Collapse
Affiliation(s)
- Zhijun Han
- Department of Thoracic Surgery, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Beijing, 100730, P.R. China
| | - Yangyang Li
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 100101, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Jian Zhang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 100101, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Chongye Guo
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 100101, P.R. China
| | - Qian Li
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 100101, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Xin Zhang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 100101, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Yongqing Lan
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 100101, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Wenbin Gu
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 100101, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Zhikai Xing
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 100101, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Liang Liang
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 100101, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| | - Meng Li
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 100101, P.R. China
| | - Shuangli Mi
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, 100101, P.R. China.,University of Chinese Academy of Sciences, Beijing, 100049, P.R. China
| |
Collapse
|
49
|
Telo S, Calderoni L, Vichi S, Zagni F, Castellucci P, Fanti S. Alternative and New Radiopharmaceutical Agents for Lung Cancer. Curr Radiopharm 2020; 13:185-194. [PMID: 31868150 PMCID: PMC8206190 DOI: 10.2174/1874471013666191223151402] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 01/27/2019] [Accepted: 11/11/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND FDG PET/CT imaging has an established role in lung cancer (LC) management. Whilst it is a sensitive technique, FDG PET/CT has a limited specificity in the differentiation between LC and benign conditions and is not capable of defining LC heterogeneity since FDG uptake varies between histotypes. OBJECTIVE To get an overview of new radiopharmaceuticals for the study of cancer biology features beyond glucose metabolism in LC. METHODS A comprehensive literature review of PubMed/Medline was performed using a combination of the following keywords: "positron emission tomography", "lung neoplasms", "non-FDG", "radiopharmaceuticals", "tracers". RESULTS Evidences suggest that proliferation markers, such as 18F-Fluorothymidine and 11CMethionine, improve LC staging and are useful in evaluating treatment response and progression free survival. 68Ga-DOTA-peptides are already routinely used in pulmonary neuroendocrine neoplasms (NENs) management and should be firstly performed in suspected NENs. 18F-Fluoromisonidazole and other radiopharmaceuticals show a promising impact on staging, prognosis assessment and therapy response in LC patients, by visualizing hypoxia and perfusion. Radiolabeled RGD-peptides, targeting angiogenesis, may have a role in LC staging, treatment outcome and therapy. PET radiopharmaceuticals tracing a specific oncogene/signal pathway, such as EGFR or ALK, are gaining interest especially for therapeutic implications. Other PET tracers, like 68Ga-PSMA-peptides or radiolabeled FAPIs, need more development in LC, though, they are promising for therapy purposes. CONCLUSION To date, the employment of most of the described tracers is limited to the experimental field, however, research development may offer innovative opportunities to improve LC staging, characterization, stratification and response assessment in an era of increased personalized therapy.
Collapse
Affiliation(s)
- Silvi Telo
- Address correspondence to this author at the Department of Metropolitan Nuclear Medicine, University of Bologna, Bologna, Italy; Tel/Fax: +390512143959; E-mail:
| | | | | | | | | | | |
Collapse
|
50
|
SNRPB promotes the tumorigenic potential of NSCLC in part by regulating RAB26. Cell Death Dis 2019; 10:667. [PMID: 31511502 PMCID: PMC6739327 DOI: 10.1038/s41419-019-1929-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/29/2019] [Accepted: 08/27/2019] [Indexed: 12/13/2022]
Abstract
SNRPB is a core component of spliceosome and plays a major role in regulating alternative splicing of the pre-mRNA. However, little is known about its role in cancer to date. In this study, we observe that SNRPB is overexpressed in NSCLC and correlated with poor prognosis in patients with NSCLC. We demonstrate that SNRPB promotes NSCLC tumorigenesis both in vitro and in vivo. Mechanistically, we reveal that RAB26 is a critical target of SNRPB. Suppression of SNRPB leads to retention of intron seven in the RAB26 mRNA and reduced RAB26 mRNA through activation of nonsense-mediated RNA decay (NMD). Moreover, forced expression of RAB26 partially restores the decreased tumorigenicity in NSCLC cells with SNRPB depletion. Our study unveils a novel role of SNRPB in facilitating NSCLC tumorigenesis via regulation of RAB26 expression and proposes that the SNRPB/RAB26 pathway may offer a therapeutic vulnerability in NSCLC.
Collapse
|