We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Fatty acid metabolism-related lncRNA prognostic signature for serous ovarian carcinoma

    Lele Ye†

    Women's Reproductive Health Laboratory of Zhejiang Province, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China

    †Authors contributed equally

    Search for more papers by this author

    ,
    Zhuofeng Jiang†

    Department of Biochemistry, School of Medicine, Southern University of Science & Technology, Shenzhen, 518055, Guangdong, China

    †Authors contributed equally

    Search for more papers by this author

    ,
    Mengxia Zheng†

    Women's Reproductive Health Laboratory of Zhejiang Province, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China

    †Authors contributed equally

    Search for more papers by this author

    ,
    Kan Pan

    First Clinical College, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China

    ,
    Jingru Lian

    Department of Biochemistry, School of Medicine, Southern University of Science & Technology, Shenzhen, 518055, Guangdong, China

    ,
    Bing Ju

    Department of Biochemistry, School of Medicine, Southern University of Science & Technology, Shenzhen, 518055, Guangdong, China

    ,
    Xuefei Liu

    Department of Biochemistry, School of Medicine, Southern University of Science & Technology, Shenzhen, 518055, Guangdong, China

    ,
    Sangsang Tang

    Women's Reproductive Health Laboratory of Zhejiang Province, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China

    ,
    Gangqiang Guo

    Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research & Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens & Immunity, Department of Microbiology & Immunology, Institute of Molecular Virology & Immunology, Institute of Tropical Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China

    ,
    Songfa Zhang

    Department of Gynecologic Oncology, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China

    ,
    Xin Hong

    **Author for correspondence:

    E-mail Address: hongx@sustech.edu.cn

    Department of Biochemistry, School of Medicine, Southern University of Science & Technology, Shenzhen, 518055, Guangdong, China

    Key University Laboratory of Metabolism & Health of Guangdong, Southern University of Science & Technology, Shenzhen, 518055, Guangdong, China

    Guangdong Provincial Key Laboratory of Cell Microenvironment & Disease Research, Southern University of Science & Technology, Shenzhen, 518055, Guangdong, China

    &
    Weiguo Lu

    *Author for correspondence:

    E-mail Address: lbwg@zju.edu.cn

    Women's Reproductive Health Laboratory of Zhejiang Province, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China

    Department of Gynecologic Oncology, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China

    Center of Uterine Cancer Diagnosis & Therapy of Zhejiang Province, Hangzhou, 310006, Zhejiang, China

    Published Online:https://doi.org/10.2217/epi-2023-0388

    Background: To explore the role of fatty acid metabolism (FAM)-related lncRNAs in the prognosis and antitumor immunity of serous ovarian cancer (SOC). Materials & methods: A SOC FAM-related lncRNA risk model was developed and evaluated by a series of analyses. Additional immune-related analyses were performed to further assess the associations between immune state, tumor microenvironment and the prognostic risk model. Results: Five lncRNAs associated with the FAM genes were found and used to create a predictive risk model. The patients with a low-risk profile exhibited favorable prognostic outcomes. Conclusion: The established prognostic risk model exhibits better predictive capabilities for the prognosis of patients with SOC and offers novel potential therapy targets for SOC.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Zheng A, Wei Y, Zhao Y, Zhang T, Ma X. The role of cancer-associated mesothelial cells in the progression and therapy of ovarian cancer. Front. Immunol. 13, 1013506 (2022).
    • 2. Barnes BM, Nelson L, Tighe A et al. Distinct transcriptional programs stratify ovarian cancer cell lines into the five major histological subtypes. Genome Med. 13(1), 140 (2021).
    • 3. Lu X, Ji C, Jiang L et al. Tumour microenvironment-based molecular profiling reveals ideal candidates for high-grade serous ovarian cancer immunotherapy. Cell Prolif. 54(3), e12979 (2021). • Revealed two immune subtypes with different responses to immunotherapy and indicated that some high-grade serous ovarian cancers may be susceptible to immunotherapies or combination therapies.
    • 4. Gong J, Chehrazi-Raffle A, Reddi S, Salgia R. Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations. J. Immunother. Cancer 6(1), 8 (2018).
    • 5. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 18(3), 197–218 (2019). • Detailed review defining hot, altered and cold tumors, the complexity of the tumor microenvironment, the Immunoscore and immune context of tumors.
    • 6. Yang C, Xia BR, Zhang ZC, Zhang YJ, Lou G, Jin WL. Immunotherapy for ovarian cancer: adjuvant, combination, and neoadjuvant. Front. Immunol. 11, 577869 (2020).
    • 7. Angell H, Galon J. From the immune contexture to the Immunoscore: the role of prognostic and predictive immune markers in cancer. Curr. Opin. Immunol. 25(2), 261–267 (2013).
    • 8. James NE, Woodman M, Ribeiro JR. Prognostic immunologic signatures in epithelial ovarian cancer. Oncogene 41(10), 1389–1396 (2022).
    • 9. Zhao H, Gu S, Bao S et al. Mechanistically derived patient-level framework for precision medicine identifies a personalized immune prognostic signature in high-grade serous ovarian cancer. Brief. Bioinform. 22(3), bbaa069 (2021).
    • 10. Karagoz K, Mehta GA, Khella CA, Khanna P, Gatza ML. Integrative proteogenomic analyses of human tumours identifies ADNP as a novel oncogenic mediator of cell cycle progression in high-grade serous ovarian cancer with poor prognosis. EBioMedicine 50, 191–202 (2019).
    • 11. Sun J, Yan C, Xu D et al. Immuno-genomic characterisation of high-grade serous ovarian cancer reveals immune evasion mechanisms and identifies an immunological subtype with a favourable prognosis and improved therapeutic efficacy. Br. J. Cancer 126(11), 1570–1580 (2022).
    • 12. Yan C, Li K, Meng F et al. Integrated immunogenomic analysis of single-cell and bulk tissue transcriptome profiling unravels a macrophage activation paradigm associated with immunologically and clinically distinct behaviors in ovarian cancer. J. Adv. Res. 44, 149–160 (2023).
    • 13. Reilly NA, Lutgens E, Kuiper J, Heijmans BT, Wouter Jukema J. Effects of fatty acids on T cell function: role in atherosclerosis. Nat. Rev. Cardiol. 18(12), 824–837 (2021). • Detailed review of the effects of fatty acids on four components of the T cell response: metabolism, activation, proliferation and polarization.
    • 14. Huang L, Gao L, Chen C. Role of medium-chain fatty acids in healthy metabolism: a clinical perspective. Trends Endocrinol. Metab. 32(6), 351–366 (2021).
    • 15. Gimple RC, Kidwell RL, Kim LJY et al. Glioma stem cell-specific superenhancer promotes polyunsaturated fatty-acid synthesis to support EGFR signaling. Cancer Discov. 9(9), 1248–1267 (2019).
    • 16. Qu YY, Zhao R, Zhang HL et al. Inactivation of the AMPK-GATA3-ECHS1 pathway induces fatty acid synthesis that promotes clear cell renal cell carcinoma growth. Cancer Res. 80(2), 319–333 (2020).
    • 17. Zhu J, Wu G, Song L et al. NKX2–8 deletion-induced reprogramming of fatty acid metabolism confers chemoresistance in epithelial ovarian cancer. EBioMedicine 43, 238–252 (2019).
    • 18. Yoon H, Lee S. Fatty acid metabolism in ovarian cancer: therapeutic implications. Int. J. Mol. Sci. 23(4), 2170 (2022).•• Detailed review of the role of fatty acid metabolism in tumor immunity; suggests that fatty acid and related lipid metabolic pathways are potential therapeutic targets for ovarian cancer.
    • 19. Xuan Y, Wang H, Yung MM et al. SCD1/FADS2 fatty acid desaturases equipoise lipid metabolic activity and redox-driven ferroptosis in ascites-derived ovarian cancer cells. Theranostics 12(7), 3534–3552 (2022).
    • 20. Cao T, Dong J, Huang J, Tang Z, Shen H. Identification of fatty acid signature to predict prognosis and guide clinical therapy in patients with ovarian cancer. Front. Oncol. 12, 979565 (2022).
    • 21. Peck B, Schulze A. Lipid metabolism at the nexus of diet and tumor microenvironment. Trends Cancer 5(11), 693–703 (2019).
    • 22. He Q, Yang C, Xiang Z et al. LINC00924-induced fatty acid metabolic reprogramming facilitates gastric cancer peritoneal metastasis via hnRNPC-regulated alternative splicing of Mnk2. Cell Death Dis. 13(11), 987 (2022).
    • 23. Jia Y, Yan Q, Zheng Y et al. Long non-coding RNA NEAT1 mediated RPRD1B stability facilitates fatty acid metabolism and lymph node metastasis via c-Jun/c-Fos/SREBP1 axis in gastric cancer. J. Exp. Clin. Cancer Res. 41(1), 287 (2022).
    • 24. Chang CY, Yang L, Tse J et al. Genetic variations in UCA1, a lncRNA functioning as a miRNA sponge, determine endometriosis development and the potential associated infertility via regulating lipogenesis. PLOS ONE 17(7), e0271616 (2022).
    • 25. Shang C, Wang W, Liao Y et al. LNMICC promotes nodal metastasis of cervical cancer by reprogramming fatty acid metabolism. Cancer Res. 78(4), 877–890 (2018). •• Established the role of a novel lncRNA in lymph node metastasis, with implications as a candidate prognostic biomarker and therapeutic target in cervical cancer.
    • 26. Lang X, Huang C, Cui H. Prognosis analysis and validation of fatty acid metabolism-related lncRNAs and tumor immune microenvironment in cervical cancer. J. Immunol. Res. 2022, 4954457 (2022).
    • 27. Wang H, Cui J, Yu J, Huang J, Li M. Identification of fatty acid metabolism-related lncRNAs as biomarkers for clinical prognosis and immunotherapy response in patients with lung adenocarcinoma. Front. Genet. 13, 855940 (2022).
    • 28. Chen E, Yi J, Jiang J et al. Identification and validation of a fatty acid metabolism-related lncRNA signature as a predictor for prognosis and immunotherapy in patients with liver cancer. BMC Cancer 22(1), 1037 (2022).
    • 29. Xu F, Huang XL, Li YY, Chen YS, Lin L. m6A-related lncRNAs are potential biomarkers for predicting prognoses and immune responses in patients with LUAD. Mol. Ther. Nucleic Acids 24, 780–791 (2021).
    • 30. Ye LL, Pan K, Fang S et al. Four types of RNA modification writer-related lncRNAs are effective predictors of prognosis and immunotherapy response in serous ovarian carcinoma. Front. Immunol. 13, 863484 (2022).
    • 31. Keum N, Giovannucci E. Global burden of colorectal cancer: emerging trends, risk factors and prevention strategies. Nat. Rev. Gastroenterol. Hepatol. 16(12), 713–732 (2019).
    • 32. Zhang Z, Zhang C, Yang Z et al. m6A regulators as predictive biomarkers for chemotherapy benefit and potential therapeutic targets for overcoming chemotherapy resistance in small-cell lung cancer. J. Hematol. Oncol. 14(1), 190 (2021).
    • 33. Subramanian A, Tamayo P, Mootha VK et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102(43), 15545–15550 (2005).
    • 34. Hanzelmann S, Castelo R, Guinney J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics 14, 7 (2013).
    • 35. Aran D, Hu Z, Butte AJ. xCell: digitally portraying the tissue cellular heterogeneity landscape. Genome Biol. 18(1), 220 (2017).
    • 36. Becht E, Giraldo NA, Lacroix L et al. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol. 17(1), 218 (2016).
    • 37. Newman AM, Liu CL, Green MR et al. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods 12(5), 453–457 (2015).
    • 38. Maeser D, Gruener RF, Huang RS. oncoPredict: an R package for predicting or cancer patient drug response and biomarkers from cell line screening data. Brief. Bioinform. 22(6), bbab260 (2021).
    • 39. Wang C, Zhang CX, Yang SM, Xiang JD, Zhou DM, Xi XW. Identification and validation of m5c-related lncRNA risk model for ovarian cancer. J. Ovarian Res. 16(1), 96 (2023).
    • 40. Geng R, Chen T, Zhong ZH, Ni SM, Bai JL, Liu JH. The m6A-related long noncoding RNA signature predicts prognosis and indicates tumor immune infiltration in ovarian cancer. Cancers 14(16), 4056 (2022).
    • 41. Zheng JF, Guo JL, Cao BB, Zhou Y, Tong JY. Identification and validation of lncRNAs involved in m6A regulation for patients with ovarian cancer. Cancer Cell Int. 21(1), 363 (2021).
    • 42. Nie X, Tan JC. N6-methyladenosine-related lncRNAs is a potential marker for predicting prognosis and immunotherapy in ovarian cancer. Hereditas 159(1), 17 (2022).
    • 43. Hu XQ, Zhang XC, Li ST, Hua T. Construction and validation of a histone acetylation-related lncRNA prognosis signature for ovarian cancer. Front. Genet. 13, 934246 (2022).
    • 44. Yang SY,J, i J, Wang M, Nie JF, Wang SJ. Construction of ovarian cancer prognostic model based on the investigation of ferroptosis-related lncRNA. Biomolecules 13(2), 306 (2023).
    • 45. Wang KL, Mei SS, Cai MC et al. Ferroptosis-related long noncoding RNAs as prognostic biomarkers for ovarian cancer. Front. Oncol. 12, 888699 (2022).
    • 46. Zheng JF, Guo JL, Wang YH, Zheng YL, Zhang K, Tong JY. Bioinformatic analyses of the ferroptosis-related lncRNAs signature for ovarian cancer. Front. Mol. Biosci. 8, 735871 (2022).
    • 47. Gao J, Pang XA, Ren F, Zhu LC. Identification of a ferroptosis-related long non-coding RNA signature for prognosis prediction of ovarian cancer. Carcinogenesis 44(1), 80–92 (2023).
    • 48. Wang YC, Liang Q, Xu L et al. Cuproptosis-related lncRNAs ovarian cancer: multi-omics analysis of molecular mechanisms and potential therapeutic targets. Environ. Toxicol. doi: 10.1002/tox.24067 (2023) (Epub ahead of print).
    • 49. Liu L, Wang Q, Zhou JY, Zhang B. Developing four cuproptosis-related lncRNAs signature to predict prognosis and immune activity in ovarian cancer. J. Ovarian Res. 16(1), 88 (2023).
    • 50. Li Y, Wang J, Wang F, Gao CZ, Cao YY, Wang JH. Development and verification of an autophagy-related lncRNA signature to predict clinical outcomes and therapeutic responses in ovarian cancer. Front. Med. 8, 715250 (2021).
    • 51. Wang XF, Dai CY, Ye MQ, Wang JY, Lin WZ, Li RM. Prognostic value of an autophagy-related long-noncoding-RNA signature for endometrial cancer. Aging 13(4), 5104–5119 (2021).
    • 52. Zhang ZY, Xu ZJ, Yan YL. Role of a pyroptosis-related lncRNA signature in risk stratification and immunotherapy of ovarian cancer. Front. Med. 8, 793515 (2022).
    • 53. He YB, Fang LW, Hu D et al. Necroptosis-associated long noncoding RNAs can predict prognosis and differentiate between cold and hot tumors in ovarian cancer. Front. Oncol. 12, 999654 (2022).
    • 54. Chen L, Gao WJ, Lin L et al. A methylation- and immune-related lncRNA signature to predict ovarian cancer outcome and uncover mechanisms of chemoresistance. J. Ovarian Res. 16(1), (2023).
    • 55. Feng J, Yu YP, Yin W, Qian SM. Development and verification of a 7-lncRNA prognostic model based on tumor immunity for patients with ovarian cancer. J. Ovarian Res. 16(1), (2023).
    • 56. Li H, Liu ZY, Chen YC, Zhang XY, Wu NYY, Wang J. Identification and validation of an immune-related lncRNAs signature to predict the overall survival of ovarian cancer. Front. Oncol. 12 (2022).
    • 57. Pan X, Bi FF. A potential immune-related long non-coding RNA prognostic signature for ovarian cancer. Front. Genet. 12, 694009 (2021).
    • 58. Sun XY, Li S, Lv XM et al. Immune-related long non-coding RNA constructs a prognostic signature of ovarian cancer. Biol. Proced. Online 23(1), 24 (2021).
    • 59. Peng Y, Wang H, Huang Q, Wu JJ, Zhang MJ. A prognostic model based on immune-related long noncoding RNAs for patients with epithelial ovarian cancer. J. Ovarian Res. 15(1), 8 (2022).
    • 60. Wang J, Zhang XC, Zheng F, Yang Q, Bi FF. Mitophagy-related long non-coding RNA signature predicts prognosis and drug response in ovarian cancer. J. Ovarian Res. 16(1), 177 (2023).
    • 61. Zheng JF, Guo JL, Zhu LL, Zhou Y, Tong JY. Comprehensive analyses of glycolysis-related lncRNAs for ovarian cancer patients. J. Ovarian Res. 14(1), 124 (2021).
    • 62. Hua T, Zhang XC, Wang W, Tian YJ, Chen SB. Deciphering the expression patterns of homologous recombination-related lncRNAs identifies new molecular subtypes and emerging therapeutic opportunities in epithelial ovarian cancer. Front. Genet. 13, 901424 (2022).
    • 63. Guo QY, He Y, Sun LY et al. Identification of potential prognostic TF-associated lncRNAs for predicting survival in ovarian cancer. J. Cell. Mol. Med. 23(3), 1840–1851 (2019).
    • 64. Xu L, Zhu S, Lan Y et al. Revealing the contribution of somatic gene mutations to shaping tumor immune microenvironment. Brief. Bioinform. 23(3), bbac064 (2022).
    • 65. Matsuda T, Leisegang M, Park JH et al. Induction of neoantigen-specific cytotoxic T cells and construction of T-cell receptor-engineered T cells for ovarian cancer. Clin. Cancer Res. 24(21), 5357–5367 (2018).
    • 66. Lang F, Schrors B, Lower M, Tureci O, Sahin U. Identification of neoantigens for individualized therapeutic cancer vaccines. Nat. Rev. Drug Discov. 21(4), 261–282 (2022).
    • 67. Yang W, Soares J, Greninger P et al. Genomics of Drug Sensitivity in Cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 41(Database issue), D955–961 (2013).
    • 68. Lheureux S, Braunstein M, Oza AM. Epithelial ovarian cancer: evolution of management in the era of precision medicine. CA Cancer J. Clin. 69(4), 280–304 (2019).
    • 69. Want MY, Lugade AA, Battaglia S, Odunsi K. Nature of tumour rejection antigens in ovarian cancer. Immunology 155(2), 202–210 (2018).
    • 70. Kurnit KC, Fleming GF, Lengyel E. Updates and new options in advanced epithelial ovarian cancer treatment. Obstet. Gynecol. 137(1), 108–121 (2021).
    • 71. Kuroki L, Guntupalli SR. Treatment of epithelial ovarian cancer. BMJ 371, m3773 (2020).
    • 72. Ray-Coquard I, Pautier P, Pignata S et al. Olaparib plus bevacizumab as first-line maintenance in ovarian cancer. N. Engl. J. Med. 381(25), 2416–2428 (2019).
    • 73. Gonzalez-Martin A, Pothuri B, Vergote I et al. Niraparib in patients with newly diagnosed advanced ovarian cancer. N. Engl. J. Med. 381(25), 2391–2402 (2019).
    • 74. Pujade-Lauraine E, Fujiwara K, Ledermann JA et al. Avelumab alone or in combination with chemotherapy versus chemotherapy alone in platinum-resistant or platinum-refractory ovarian cancer (JAVELIN Ovarian 200): an open-label, three-arm, randomised, phase 3 study. Lancet Oncol. 22(7), 1034–1046 (2021).
    • 75. Vivot A, Crequit P, Porcher R. Use of late-life expectancy for assessing the long-term benefit of immune checkpoint inhibitors. J. Natl Cancer Inst. 111(5), 519–521 (2019).
    • 76. Lheureux S, Gourley C, Vergote I, Oza AM. Epithelial ovarian cancer. Lancet 393(10177), 1240–1253 (2019).
    • 77. He W, Liang B, Wang C et al. MSC-regulated lncRNA MACC1-AS1 promotes stemness and chemoresistance through fatty acid oxidation in gastric cancer. Oncogene 38(23), 4637–4654 (2019).
    • 78. Cui M, Xiao Z, Wang Y et al. Long noncoding RNA HULC modulates abnormal lipid metabolism in hepatoma cells through an miR-9-mediated RXRA signaling pathway. Cancer Res. 75(5), 846–857 (2015).
    • 79. Fang H, Li H, Zhang H et al. Short-chain L-3-hydroxyacyl-CoA dehydrogenase: a novel vital oncogene or tumor suppressor gene in cancers. Front. Pharmacol. 13, 1019312 (2022).
    • 80. Motohara T, Masuda K, Morotti M et al. An evolving story of the metastatic voyage of ovarian cancer cells: cellular and molecular orchestration of the adipose-rich metastatic microenvironment. Oncogene 38(16), 2885–2898 (2019).
    • 81. Park EG, Pyo SJ, Cui Y, Yoon SH, Nam JW. Tumor immune microenvironment lncRNAs. Brief. Bioinform. 23(1), bbab504 (2022). •• Detailed review of the regulatory functions of the lncRNAs at the cell type level; highlights how a scRNA-sequencing approach can help to study the cell type-specific functions of tumor immune microenvironment lncRNAs.
    • 82. Zhu L, Zhu X, Wu Y. Effects of glucose metabolism, lipid metabolism, and glutamine metabolism on tumor microenvironment and clinical implications. Biomolecules 12(4), 580 (2022).
    • 83. Roche J. The epithelial-to-mesenchymal transition in cancer. Cancers (Basel) 10(2), 52 (2018).
    • 84. Kao KC, Vilbois S, Tsai CH, Ho PC. Metabolic communication in the tumour-immune microenvironment. Nat. Cell Biol. 24(11), 1574–1583 (2022).
    • 85. Corn KC, Windham MA, Rafat M. Lipids in the tumor microenvironment: from cancer progression to treatment. Prog. Lipid Res. 80, 101055 (2020).
    • 86. Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 14(10), 1014–1022 (2013).
    • 87. Schumacher TN, Thommen DS. Tertiary lymphoid structures in cancer. Science 375(6576), eabf9419 (2022).
    • 88. Li K, Shi H, Zhang B et al. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduct. Target. Ther. 6(1), 362 (2021).
    • 89. Knochelmann HM, Dwyer CJ, Bailey SR et al. When worlds collide: Th17 and Treg cells in cancer and autoimmunity. Cell. Mol. Immunol. 15(5), 458–469 (2018).
    • 90. Pan Y, Yu Y, Wang X, Zhang T. Tumor-associated macrophages in tumor immunity. Front. Immunol. 11, 583084 (2020).
    • 91. Chen YJ, Li GN, Li XJ et al. Targeting IRG1 reverses the immunosuppressive function of tumor-associated macrophages and enhances cancer immunotherapy. Sci. Adv. 9(17), eadg0654 (2023).
    • 92. Addeo A, Friedlaender A, Banna GL, Weiss GJ. TMB or not TMB as a biomarker: that is the question. Crit. Rev. Oncol. Hematol. 163, 103374 (2021).
    • 93. Jardim DL, Goodman A, De Melo Gagliato D, Kurzrock R. The challenges of tumor mutational burden as an immunotherapy biomarker. Cancer Cell 39(2), 154–173 (2021).
    • 94. Bieging KT, Mello SS, Attardi LD. Unravelling mechanisms of p53-mediated tumour suppression. Nat. Rev. Cancer 14(5), 359–370 (2014).
    • 95. Muller PA, Vousden KH. Mutant p53 in cancer: new functions and therapeutic opportunities. Cancer Cell 25(3), 304–317 (2014).
    • 96. Freed-Pastor WA, Prives C. Mutant p53: one name, many proteins. Genes Dev. 26(12), 1268–1286 (2012).
    • 97. Ren YA, Mullany LK, Liu Z, Herron AJ, Wong KK, Richards JS. Mutant p53 promotes epithelial ovarian cancer by regulating tumor differentiation, metastasis, and responsiveness to steroid hormones. Cancer Res. 76(8), 2206–2218 (2016).
    • 98. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N. Engl. J. Med. 377(25), 2500–2501 (2017).
    • 99. Hellmann MD, Nathanson T, Rizvi H et al. Genomic features of response to combination immunotherapy in patients with advanced non-small-cell lung cancer. Cancer Cell 33(5), 843–852; e844 (2018).• Revealed the association between tumor mutation burden and benefit to combination immunotherapy in non-small-cell lung cancer.
    • 100. Rizvi H, Sanchez-Vega F, La K et al. Molecular determinants of response to anti-programmed cell death (PD)-1 and anti-programmed death-ligand 1 (PD-L1) blockade in patients with non-small-cell lung cancer profiled with targeted next-generation sequencing. J. Clin. Oncol. 36(7), 633–641 (2018).
    • 101. Hellmann MD, Callahan MK, Awad MM et al. Tumor mutational burden and efficacy of nivolumab monotherapy and in combination with ipilimumab in small-cell lung cancer. Cancer Cell 33(5), 853–861; e854 (2018).
    • 102. Fan S, Gao X, Qin Q, Li H, Yuan Z, Zhao S. Association between tumor mutation burden and immune infiltration in ovarian cancer. Int. Immunopharmacol. 89(Pt A), 107126 (2020).
    • 103. Goodman AM, Piccioni D, Kato S et al. Prevalence of PDL1 amplification and preliminary response to immune checkpoint blockade in solid tumors. JAMA Oncol. 4(9), 1237–1244 (2018).