Article Data

  • Views 906
  • Dowloads 120

Original Research

Open Access

Construction of a ferroptosis-related lncRNA signature for predicting prognosis, immune response and drug sensitivity in cervical squamous cell carcinoma and endocervical adenocarcinoma

  • Tingting Gu1,†
  • Caihong Xu2,†
  • Jing Chen2
  • Bin Wan2
  • Wei Wang3,*,
  • Jun Shi1,4,*,

1Department of Obstetrics and Gynecology, Affiliated Hospital of Nanjing University of Chinese Medicine, Taicang Hospital of Traditional Chinese Medicine, 215400 Taicang, Jiangsu, China

2Department of Obstetrics and Gynecology, Nanjing Tongren Hospital, School of Medicine, Southeast University, 211102 Nanjing, Jiangsu, China

3Department of Clinical Laboratory, Lianshui County People’s Hospital, 223400 Huai’an, Jiangsu, China

4Department of Clinical Laboratory, Affiliated Hospital of Nanjing University of Chinese Medicine, Taicang Hospital of Traditional Chinese Medicine, 215400 Taicang, Jiangsu, China

DOI: 10.22514/ejgo.2023.096 Vol.44,Issue 6,December 2023 pp.29-42

Submitted: 17 February 2023 Accepted: 25 March 2023

Published: 15 December 2023

*Corresponding Author(s): Wei Wang E-mail: wangwei_shzu@163.com
*Corresponding Author(s): Jun Shi E-mail: sj1987517@163.com

† These authors contributed equally.

Abstract

Ferroptosis, a recently identified cell death mode, has been shown to play critical roles in several malignant tumors. Long non-coding RNAs (lncRNAs) have been reported to modulate ferroptosis, thereby affecting the growth and prognosis of cancers. However, the association between lncRNA and ferroptosis in cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) remains unclear. The aim of this study was to identify ferroptosis-related lncRNAs (FRLs) associated with CESC prognosis and investigate their interaction with tumor immune response. R software and Perl were used to screen for aberrant expressed FRLs associated with CESC patient prognosis from The Cancer Genome Atlas (TCGA) database, including AP003774.2, SOX21 antisense divergent transcript 1 (SOX21-AS1), myocardial infarction associated transcript (MIAT), RUSC1 antisense RNA 1 (RUSC1-AS1), AC004847.1, AC009097.2, MIR100 host gene (MIR100HG), AC083799.1, long intergenic non-protein coding RNA 958 (LINC00958), AC009065.8 and AC131159.1. A risk score was calculated for each CESC patient individual according to the expression levels of 11 FRLs, based on which a prognostic model was built. Kaplan-Meier (K-M) survival curve analysis and Receiver Operating Characteristic (ROC) curve assessment were conducted to determine the predictive accuracy of the prognostic model. Lastly, the R software and CIBERSORT were employed to examine the differences in immune cell infiltration, immune checkpoint and drug sensitivity between the two subgroups. The 11 FRLs were used to construct a prognostic model that classified CESC patients into a high- or low-risk group. The FRL-based model was found to outperform traditional clinicopathological features in predicting CESC patient survival. Significant variations existed across subgroups in immune cell infiltration, immunological function, overall survival (OS), and inhibitory concentrations (IC50 values). Our findings provide novel insights into the role of FRLs in CESC and present a personalized predictive tool for determining patient prognosis, immune response and drug sensitivity.


Keywords

Ferroptosis; Immune cell infiltrate; Drug sensitivity; Biomarkers; LncRNA; Survival analysis; Cervical squamous cell carcinoma and endocervical adenocarcinoma


Cite and Share

Tingting Gu,Caihong Xu,Jing Chen,Bin Wan,Wei Wang,Jun Shi. Construction of a ferroptosis-related lncRNA signature for predicting prognosis, immune response and drug sensitivity in cervical squamous cell carcinoma and endocervical adenocarcinoma. European Journal of Gynaecological Oncology. 2023. 44(6);29-42.

References

[1] Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: A Cancer Journal for Clinicians. 2021; 71: 209–249.

[2] Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA: A Cancer Journal for Clinicians. 2022; 72: 7–33.

[3] Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012; 149: 1060–1072.

[4] Tang R, Hua J, Xu J, Liang C, Meng Q, Liu J, et al. The role of ferroptosis regulators in the prognosis, immune activity and gemcitabine resistance of pancreatic cancer. Annals of Translational Medicine. 2020; 8: 1347.

[5] Yang WS, Stockwell BR. Ferroptosis: death by lipid peroxidation. Trends in Cell Biology. 2016; 26: 165–176.

[6] Chen X, Kang R, Kroemer G, Tang D. Broadening horizons: the role of ferroptosis in cancer. Nature Reviews Clinical Oncology. 2021; 18: 280–296.

[7] Fan F, Liu P, Bao R, Chen J, Zhou M, Mo Z, et al. A dual PI3K/HDAC inhibitor induces immunogenic ferroptosis to potentiate cancer immune checkpoint therapy. Cancer Research. 2021; 81: 6233–6245.

[8] Chen X, Kang R, Kroemer G, Tang D. Ferroptosis in infection, inflammation, and immunity. Journal of Experimental Medicine. 2021; 218: e20210518.

[9] Dodson M, Castro-Portuguez R, Zhang DD. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biology. 2019; 23: 101107.

[10] Ouyang S, Li H, Lou L, Huang Q, Zhang Z, Mo J, et al. Inhibition of STAT3-ferroptosis negative regulatory axis suppresses tumor growth and alleviates chemoresistance in gastric cancer. Redox Biology. 2022; 52: 102317.

[11] Wei R, Zhao Y, Wang J, Yang X, Li S, Wang Y, et al. Tagitinin C induces ferroptosis through PERK-Nrf2-HO-1 signaling pathway in colorectal cancer cells. International Journal of Biological Sciences. 2021; 17: 2703–2717.

[12] Shan G, Zhang H, Bi G, Bian Y, Liang J, Valeria B, et al. Multi-omics analysis of cancer cell lines with high/low ferroptosis scores and development of a ferroptosis-related model for multiple cancer types. Frontiers in Cell and Developmental Biology. 2021; 9: 794475.

[13] Wang W, Zhang J, Wang Y, Xu Y, Zhang S. Identifies microtubule-binding protein CSPP1 as a novel cancer biomarker associated with ferroptosis and tumor microenvironment. Computational and Structural Biotechnology Journal. 2022; 20: 3322–3335.

[14] Xing C, Yin H, Yao ZY, Xing XL. Prognostic signatures based on ferroptosis- and immune-related genes for cervical squamous cell carcinoma and endocervical adenocarcinoma. Frontiers in Oncology. 2021; 11: 774558.

[15] Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, et al. Landscape of transcription in human cells. Nature. 2012; 489: 101–108.

[16] Rinn JL, Chang HY. Genome regulation by long noncoding RNAs. Annual Review of Biochemistry. 2012; 81: 145–166.

[17] He T, Yuan C, Zhao C. Long intragenic non-coding RNA p53-induced transcript (LINC-PINT) as a novel prognosis indicator and therapeutic target in cancer. Biomedicine & Pharmacotherapy. 2021; 143: 112127.

[18] Kong X, Duan Y, Sang Y, Li Y, Zhang H, Liang Y, et al. LncRNA-CDC6 promotes breast cancer progression and function as ceRNA to target CDC6 by sponging microRNA-215. Journal of Cellular Physiology. 2019; 234: 9105–9117.

[19] Marín-Béjar O, Mas AM, González J, Martinez D, Athie A, Morales X, et al. The human lncRNA LINC-PINT inhibits tumor cell invasion through a highly conserved sequence element. Genome Biology. 2017; 18: 202.

[20] Wang J, Su Z, Lu S, Fu W, Liu Z, Jiang X, et al. LncRNA HOXA-as2 and its molecular mechanisms in human cancer. Clinica Chimica Acta. 2018; 485: 229–233.

[21] Zhang B, Bao W, Zhang S, Chen B, Zhou X, Zhao J, et al. LncRNA HEPFAL accelerates ferroptosis in hepatocellular carcinoma by regulating SLC7A11 ubiquitination. Cell Death & Disease. 2022; 13: 734.

[22] Zhang Y, Luo M, Cui X, O’Connell D, Yang Y. Long noncoding RNA NEAT1 promotes ferroptosis by modulating the miR-362-3p/MIOX axis as a ceRNA. Cell Death & Differentiation. 2022; 29: 1850–1863.

[23] Miller KD, Nogueira L, Devasia T, Mariotto AB, Yabroff KR, Jemal A, et al. Cancer treatment and survivorship statistics, 2022. CA: A Cancer Journal for Clinicians. 2022; 72: 409–436.

[24] Siegel R, DeSantis C, Virgo K, Stein K, Mariotto A, Smith T, et al. Cancer treatment and survivorship statistics, 2012. CA: A Cancer Journal for Clinicians. 2012; 62: 220–241.

[25] Brisson M, Kim JJ, Canfell K, Drolet M, Gingras G, Burger EA, et al. Impact of HPV vaccination and cervical screening on cervical cancer elimination: a comparative modelling analysis in 78 low-income and lower-middle-income countries. The Lancet. 2020; 395: 575–590.

[26] Lin C, Slama J, Gonzalez P, Goodman MT, Xia N, Kreimer AR, et al. Cervical determinants of anal HPV infection and high-grade anal lesions in women: a collaborative pooled analysis. The Lancet Infectious Diseases. 2019; 19: 880–891.

[27] Malla R, Kamal MA. E6 and E7 oncoproteins: potential targets of cervical cancer. Current Medicinal Chemistry. 2021; 28: 8163–8181.

[28] Colli LM, Machiela MJ, Zhang H, Myers TA, Jessop L, Delattre O, et al. Landscape of combination immunotherapy and targeted therapy to improve cancer management. Cancer Research. 2017; 77: 3666–3671.

[29] Lee YT, Tan YJ, Oon CE. Molecular targeted therapy: treating cancer with specificity. European Journal of Pharmacology. 2018; 834: 188–196.

[30] Li H, Wu X, Cheng X. Advances in diagnosis and treatment of metastatic cervical cancer. Journal of Gynecologic Oncology. 2016; 27: e34.

[31] Mun EJ, Babiker HM, Weinberg U, Kirson ED, Von Hoff DD. Tumor-treating fields: a fourth modality in cancer treatment. Clinical Cancer Research. 2018; 24: 266–275.

[32] Bhattacharya B, Mohd Omar MF, Soong R. The Warburg effect and drug resistance. British Journal of Pharmacology. 2016; 173: 970–979.

[33] Friedmann Angeli JP, Krysko DV, Conrad M. Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nature Reviews Cancer. 2019; 19: 405–414.

[34] Vasan N, Baselga J, Hyman DM. A view on drug resistance in cancer. Nature. 2019; 575: 299–309.

[35] Koren E, Fuchs Y. Modes of regulated cell death in cancer. Cancer Discovery. 2021; 11: 245–265.

[36] Liu P, Wang W, Li Z, Li Y, Yu X, Tu J, et al. Ferroptosis: a new regulatory mechanism in osteoporosis. Oxidative Medicine and Cellular Longevity. 2022; 2022: 1–10.

[37] Gao W, Wang X, Zhou Y, Wang X, Yu Y. Autophagy, ferroptosis, pyroptosis, and necroptosis in tumor immunotherapy. Signal Transduction and Targeted Therapy. 2022; 7: 196.

[38] Chen Q, Wang W, Wu Z, Chen S, Chen X, Zhuang S, et al. Over-expression of lncRNA TMEM161B-AS1 promotes the malignant biological behavior of glioma cells and the resistance to temozolomide via up-regulating the expression of multiple ferroptosis-related genes by sponging hsa-miR-27a-3p. Cell Death Discovery. 2021; 7: 311.

[39] Fu H, Zhang Z, Li D, Lv Q, Chen S, Zhang Z, et al. LncRNA PELATON, a ferroptosis suppressor and prognositic signature for GBM. Frontiers in oncology. 2022; 12: 817737.

[40] Huang G, Xiang Z, Wu H, He Q, Dou R, Lin Z, et al. The lncRNA BDNF-as/WDR5/FBXW7 axis mediates ferroptosis in gastric cancer peritoneal metastasis by regulating VDAC3 ubiquitination. International Journal of Biological Sciences. 2022; 18: 1415–1433.

[41] Ni T, Huang X, Pan S, Lu Z. Inhibition of the long non-coding RNA ZFAS1 attenuates ferroptosis by sponging miR-150-5p and activates CCND2 against diabetic cardiomyopathy. Journal of Cellular and Molecular Medicine. 2021; 25: 9995–10007.

[42] Mao C, Wang X, Liu Y, Wang M, Yan B, Jiang Y, et al. A G3BP1-interacting lncRNA promotes ferroptosis and apoptosis in cancer via nuclear sequestration of p53. Cancer Research. 2018; 78: 3484–3496.

[43] Luo W, Wang J, Xu W, Ma C, Wan F, Huang Y, et al. LncRNA RP11-89 facilitates tumorigenesis and ferroptosis resistance through PROM2-activated iron export by sponging miR-129-5p in bladder cancer. Cell Death & Disease. 2021; 12: 1043.

[44] Yang H, Hu Y, Weng M, Liu X, Wan P, Hu Y, et al. Hypoxia inducible lncRNA-CBSLR modulates ferroptosis through m6a-YTHDF2-dependent modulation of CBS in gastric cancer. Journal of Advanced Research. 2022; 37: 91–106.

[45] Derynck R, Muthusamy BP, Saeteurn KY. Signaling pathway cooperation in TGF-β-induced epithelial-mesenchymal transition. Current Opinion in Cell Biology. 2014; 31: 56–66.

[46] Hartsough MT, Mulder KM. Transforming growth factor-beta signaling in epithelial cells. Pharmacology & Therapeutics. 1997; 75: 21–41.

[47] Katsuno Y, Derynck R. Epithelial plasticity, epithelial-mesenchymal transition, and the TGF-β family. Developmental Cell. 2021; 56: 726–746.

[48] Sharkey DJ, Macpherson AM, Tremellen KP, Mottershead DG, Gilchrist RB, Robertson SA. TGF-β mediates proinflammatory seminal fluid signaling in human cervical epithelial cells. The Journal of Immunology. 2012; 189: 1024–1035.

[49] Xu J, Lamouille S, Derynck R. TGF-beta-induced epithelial to mesenchymal transition. Cell Research. 2009; 19: 156–172.

[50] Yue J, Mulder KM. Transforming growth factor-beta signal transduction in epithelial cells. Pharmacology & Therapeutics. 2001; 91: 1–34.

[51] Zavadil J, Böttinger EP. TGF-beta and epithelial-to-mesenchymal transitions. Oncogene. 2005; 24: 5764–5774.

[52] Wu HS, Li YF, Chou CI, Yuan CC, Hung MW, Tsai LC. The concentration of serum transforming growth factor beta-1 (TGF-beta1) is decreased in cervical carcinoma patients. Cancer Investigation. 2002; 20: 55–59.

[53] Meredith JE, Fazeli B, Schwartz MA. The extracellular matrix as a cell survival factor. Molecular Biology of the Cell. 1993; 4: 953–961.

[54] Frisch S, Francis H. Disruption of epithelial cell-matrix interactions induces apoptosis. Journal of Cell Biology. 1994; 124: 619–626.

[55] Brown CW, Amante JJ, Goel HL, Mercurio AM. The α6β4 integrin promotes resistance to ferroptosis. Journal of Cell Biology. 2017; 216: 4287–4297.

[56] Gong C, Ji Q, Wu M, Tu Z, Lei K, Luo M, et al. Ferroptosis in tumor immunity and therapy. Journal of Cellular and Molecular Medicine. 2022; 26: 5565–5579.

[57] Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nature Reviews Molecular Cell Biology. 2021; 22: 266–282.

[58] Tang R, Xu J, Zhang B, Liu J, Liang C, Hua J, et al. Ferroptosis, necroptosis, and pyroptosis in anticancer immunity. Journal of Hematology & Oncology. 2020; 13: 110.

[59] Wiernicki B, Maschalidi S, Pinney J, Adjemian S, Vanden Berghe T, Ravichandran KS, et al. Cancer cells dying from ferroptosis impede dendritic cell-mediated anti-tumor immunity. Nature Communications. 2022; 13: 3676.

[60] Xu H, Ye D, Ren M, Zhang H, Bi F. Ferroptosis in the tumor microenvironment: perspectives for immunotherapy. Trends in Molecular Medicine. 2021; 27: 856–867.

[61] Wang W, Green M, Choi JE, Gijón M, Kennedy PD, Johnson JK, et al. CD8+ T cells regulate tumour ferroptosis during cancer immunotherapy. Nature. 2019; 569: 270–274.

[62] Franzin R, Netti GS, Spadaccino F, Porta C, Gesualdo L, Stallone G, et al. The use of immune checkpoint inhibitors in oncology and the occurrence of AKI: where do we stand? Frontiers in Immunology. 2020; 11: 574271.

[63] Galluzzi L, Humeau J, Buqué A, Zitvogel L, Kroemer G. Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nature Reviews Clinical Oncology. 2020; 17: 725–741.

[64] Heinhuis KM, Ros W, Kok M, Steeghs N, Beijnen JH, Schellens JHM. Enhancing antitumor response by combining immune checkpoint inhibitors with chemotherapy in solid tumors. Annals of Oncology. 2019; 30: 219–235.

[65] Kalbasi A, Ribas A. Tumour-intrinsic resistance to immune checkpoint blockade. Nature Reviews Immunology. 2020; 20: 25–39.

[66] Miller JFAP, Sadelain M. The journey from discoveries in fundamental immunology to cancer immunotherapy. Cancer Cell. 2015; 27: 439–449.

[67] Ho P, Kaech SM. Reenergizing T cell anti-tumor immunity by harnessing immunometabolic checkpoints and machineries. Current Opinion in Immunology. 2017; 46: 38–44.

[68] Hosseinkhani N, Derakhshani A, Kooshkaki O, Abdoli Shadbad M, Hajiasgharzadeh K, Baghbanzadeh A, et al. Immune checkpoints and CAR-T cells: the pioneers in future cancer therapies? International Journal of Molecular Sciences. 2020; 21: 8305.

[69] Lei Q, Wang D, Sun K, Wang L, Zhang Y. Resistance mechanisms of anti-PD1/PDL1 therapy in solid tumors. Frontiers in Cell and Developmental Biology. 2020; 8: 672.

[70] Brahmer JR, Tykodi SS, Chow LQM, Hwu W, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. The New England Journal of Medicine. 2012; 366: 2455–2465.

[71] Daassi D, Mahoney KM, Freeman GJ. The importance of exosomal PDL1 in tumour immune evasion. Nature Reviews Immunology. 2020; 20: 209–215.

[72] Gou Q, Dong C, Xu H, Khan B, Jin J, Liu Q, et al. PD-L1 degradation pathway and immunotherapy for cancer. Cell Death & Disease. 2020; 11: 955.

[73] Jiang Q, Huang J, Zhang B, Li X, Chen X, Cui B, et al. Efficacy and safety of anti-PD1/PDL1 in advanced biliary tract cancer: a systematic review and meta-analysis. Frontiers in Immunology. 2022; 13: 801909.

[74] Jin Y, Wei J, Weng Y, Feng J, Xu Z, Wang P, et al. Adjuvant therapy with PD1/PDL1 inhibitors for human cancers: a systematic review and meta-analysis. Frontiers in Oncology. 2022; 12: 732814.

[75] Liu J, Chen Z, Li Y, Zhao W, Wu J, Zhang Z. PD-1/PD-L1 checkpoint inhibitors in tumor immunotherapy. Frontiers in Pharmacology. 2021; 12: 731798.

[76] Marchetti A, Di Lorito A, Buttitta F. Why anti-PD1/PDL1 therapy is so effective? Another piece in the puzzle. Journal of Thoracic Disease. 2017; 9: 4863–4866.

[77] Patel JJ, Levy DA, Nguyen SA, Knochelmann HM, Day TA. Impact of PD-L1 expression and human papillomavirus status in anti-PD1/PDL1 immunotherapy for head and neck squamous cell carcinoma-systematic review and meta-analysis. Head & Neck. 2020; 42: 774–786.

[78] Stenehjem DD, Tran D, Nkrumah MA, Gupta S. PD1/PDL1 inhibitors for the treatment of advanced urothelial bladder cancer. OncoTargets and Therapy. 2018; 11: 5973–5989.


Abstracted / indexed in

Science Citation Index Expanded (SciSearch) Created as SCI in 1964, Science Citation Index Expanded now indexes over 9,500 of the world’s most impactful journals across 178 scientific disciplines. More than 53 million records and 1.18 billion cited references date back from 1900 to present.

Biological Abstracts Easily discover critical journal coverage of the life sciences with Biological Abstracts, produced by the Web of Science Group, with topics ranging from botany to microbiology to pharmacology. Including BIOSIS indexing and MeSH terms, specialized indexing in Biological Abstracts helps you to discover more accurate, context-sensitive results.

Google Scholar Google Scholar is a freely accessible web search engine that indexes the full text or metadata of scholarly literature across an array of publishing formats and disciplines.

JournalSeek Genamics JournalSeek is the largest completely categorized database of freely available journal information available on the internet. The database presently contains 39226 titles. Journal information includes the description (aims and scope), journal abbreviation, journal homepage link, subject category and ISSN.

Current Contents - Clinical Medicine Current Contents - Clinical Medicine provides easy access to complete tables of contents, abstracts, bibliographic information and all other significant items in recently published issues from over 1,000 leading journals in clinical medicine.

BIOSIS Previews BIOSIS Previews is an English-language, bibliographic database service, with abstracts and citation indexing. It is part of Clarivate Analytics Web of Science suite. BIOSIS Previews indexes data from 1926 to the present.

Journal Citation Reports/Science Edition Journal Citation Reports/Science Edition aims to evaluate a journal’s value from multiple perspectives including the journal impact factor, descriptive data about a journal’s open access content as well as contributing authors, and provide readers a transparent and publisher-neutral data & statistics information about the journal.

Submission Turnaround Time

Conferences

Top