Novel cellular senescence-related risk model identified as the prognostic biomarkers for lung squamous cell carcinoma

被引:4
作者
Hu, Xiaoshan [1 ]
Guo, Liyi [2 ]
Liu, Guihong [3 ]
Dai, Zili [4 ]
Wang, Li [4 ]
Zhang, Jian [4 ,5 ]
Wang, Jun [6 ]
机构
[1] Guangzhou Med Univ, Dept Internal Med Oncol, Affiliated Canc Hosp & Inst, Guangzhou, Peoples R China
[2] Southern Med Univ, Peoples Hosp Huizhou City 6, Dept Oncol & Hematol, Huiyang Hosp, Huizhou, Peoples R China
[3] Donguan Tungwah Hosp, Dept Radiat Oncol, Dongguan, Peoples R China
[4] Guangzhou Med Univ, Guangzhou Inst Resp Dis, Dept Radiat Oncol, State Key Lab Resp Dis,Affiliated Canc Hosp & Inst, Guangzhou, Peoples R China
[5] Guangzhou Med Univ, Guangzhou, Peoples R China
[6] Guangzhou Med Univ, Dept Intervent Radiol, Affiliated Canc Hosp & Inst, Guangzhou, Peoples R China
来源
FRONTIERS IN ONCOLOGY | 2022年 / 12卷
基金
中国国家自然科学基金;
关键词
senescence; immune microenvironment; immunotherapy; lung squamous cell carcinoma; overall survival; ONCOGENE-INDUCED SENESCENCE; DNA-DAMAGE; CANCER; FIBROSIS; DISEASE;
D O I
10.3389/fonc.2022.997702
中图分类号
R73 [肿瘤学];
学科分类号
100214 ;
摘要
BackgroundLung cancer is one of the top causes of cancer-related death worldwide. Cellular senescence is a characteristic of cell cycle arrest that plays a role in carcinogenesis and immune microenvironment modulation. Despite this, the clinical and immune cell infiltration features of senescence in lung squamous cell carcinoma (LUSC) are unknown. MethodsThe Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) were used to get RNA-seq data and clinical information for LUSC. The least absolute shrinkage and selection operator (LASSO)-Cox regression, receiver operating characteristic (ROC), and Kaplan-Meier analysis were used to evaluate a risk model for predicting overall survival based on six differentially expressed genes. The tumor microenvironment (TME) and immunotherapy response were also studied. ResultsTo discriminate LUSC into high- and low-risk subgroups, a risk model comprised of six cellular senescence-related genes (CDKN1A, CEBPB, MDH1, SIX1, SNAI1, and SOX5) was developed. The model could stratify patients into high-risk and low-risk groups, according to ROC and Kaplan-Meier analysis. In the TCGA-LUSC and GSE73403 cohorts, the high-risk group had a worse prognosis (P<0.05), and was associated with immune cell inactivation and being insensitive to immunotherapy in IMvigor210. ConclusionsWe discovered a new LUSC classification based on six cellular senescence-related genes, which will aid in identifying patients who will benefit from anti-PD-1 treatment. Targeting senescence-related genes appears to be another option for improving clinical therapy for LUSC.
引用
收藏
页数:12
相关论文
共 53 条
  • [1] The homeoprotein SIX1 controls cellular senescence through the regulation of p16INK4A and differentiation-related genes
    Adrados, I.
    Larrasa-Alonso, J.
    Galarreta, A.
    Lopez-Antona, I.
    Menendez, C.
    Abad, M.
    Gil, J.
    Moreno-Bueno, G.
    Palmero, I.
    [J]. ONCOGENE, 2016, 35 (27) : 3485 - 3494
  • [2] Lopez-Dominguez JA, 2021, AGING-US, V13, P13380, DOI 10.18632/aging.203110
  • [3] Genome hyperevolution and the success of a parasite
    Barry, J. David
    Hall, James P. J.
    Plenderleith, Lindsey
    [J]. EFFECTS OF GENOME STRUCTURE AND SEQUENCE ON VARIATION AND EVOLUTION, 2012, 1267 : 11 - 17
  • [4] Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints
    Bartkova, Jirina
    Rezaei, Nousin
    Liontos, Michalis
    Karakaidos, Panagiotis
    Kletsas, Dimitris
    Issaeva, Natalia
    Vassiliou, Leandros-Vassilios F.
    Kolettas, Evangelos
    Niforou, Katerina
    Zoumpourlis, Vassilis C.
    Takaoka, Munenori
    Nakagawa, Hiroshi
    Tort, Frederic
    Fugger, Kasper
    Johansson, Fredrik
    Sehested, Maxwell
    Andersen, Claus L.
    Dyrskjot, Lars
    Orntoft, Torben
    Lukas, Jiri
    Kittas, Christos
    Helleday, Thomas
    Halazonetis, Thanos D.
    Bartek, Jiri
    Gorgoulis, Vassilis G.
    [J]. NATURE, 2006, 444 (7119) : 633 - 637
  • [5] Snail regulates cell survival and inhibits cellular senescence in human metastatic prostate cancer cell lines
    Baygi, Modjtaba Emadi
    Soheili, Zahra Soheila
    Schmitz, Ingo
    Sameie, Shahram
    Schulz, Wolfgang A.
    [J]. CELL BIOLOGY AND TOXICOLOGY, 2010, 26 (06) : 553 - 567
  • [6] Quantitative identification of senescent cells in aging and disease
    Biran, Anat
    Zada, Lior
    Abou Karam, Paula
    Vadai, Ezra
    Roitman, Lior
    Ovadya, Yossi
    Porat, Ziv
    Krizhanovsky, Valery
    [J]. AGING CELL, 2017, 16 (04): : 661 - 671
  • [7] Telomere shortening and tumor formation by mouse cells lacking telomerase RNA
    Blasco, MA
    Lee, HW
    Hande, MP
    Samper, E
    Lansdorp, PM
    DePinho, RA
    Greider, CW
    [J]. CELL, 1997, 91 (01) : 25 - 34
  • [8] Oncogene-induced senescence: Putting the brakes on tumor development
    Braig, M
    Schmitt, CA
    [J]. CANCER RESEARCH, 2006, 66 (06) : 2881 - 2884
  • [9] T-helper-1-cell cytokines drive cancer into senescence
    Braumueller, Heidi
    Wieder, Thomas
    Brenner, Ellen
    Assmann, Sonja
    Hahn, Matthias
    Alkhaled, Mohammed
    Schilbach, Karin
    Essmann, Frank
    Kneilling, Manfred
    Griessinger, Christoph
    Ranta, Felicia
    Ullrich, Susanne
    Mocikat, Ralph
    Braungart, Kilian
    Mehra, Tarun
    Fehrenbacher, Birgit
    Berdel, Julia
    Niessner, Heike
    Meier, Friedegund
    van den Broek, Maries
    Haering, Hans-Ulrich
    Handgretinger, Rupert
    Quintanilla-Martinez, Leticia
    Fend, Falko
    Pesic, Marina
    Bauer, Juergen
    Zender, Lars
    Schaller, Martin
    Schulze-Osthoff, Klaus
    Roecken, Martin
    [J]. NATURE, 2013, 494 (7437) : 361 - 365
  • [10] Cellular senescence: when bad things happen to good cells
    Campisi, Judith
    di Fagagna, Fabrizio d'Adda
    [J]. NATURE REVIEWS MOLECULAR CELL BIOLOGY, 2007, 8 (09) : 729 - 740