A metabolomic and proteomic study to elucidate the molecular mechanisms of immunotherapy resistance in patients with oesophageal squamous cell carcinoma

Lijuan Gao, Yongshun Chen, Lijuan Gao, Yongshun Chen

Abstract

Systemic chemotherapy, the standard first-line treatment option for patients with advanced oesophageal squamous cell carcinoma (OSCC), results in a median survival of ~1 year. Immune checkpoint inhibitors are a breakthrough oncology treatment option; however, most patients with advanced OSCC develop primary and acquired resistance to programmed death receptor-1 (PD-1) monoclonal antibody, severely affecting their prognosis. Therefore, there is an urgent need to investigate the molecular mechanism underlying resistance to treatment. The present study aimed to explore the mechanism of resistance to PD-1 monoclonal antibody. Plasma samples were collected from patients with OSCC treated with immunotherapy, who achieved pathological response/partial response (CR/PR) or stable disease/progressive disease (SD/PD) after the fourth treatment cycle. TM-widely targeted metabolomics, widely targeted lipidomics, and DIA proteomics assays were performed. Differential metabolites were screened based on fold change (FC) ≥1.5 or ≤0.67 and a VIP ≥1; differential proteins were screened based on FC >1.5 or <0.67 and P<0.05. The identified metabolites were annotated and mapped using the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway databases. The differential proteins were annotated to the Gene Ontology and KEGG pathway databases. A correlation network diagram was drawn using differential expressed proteins and metabolites with (Pearson correlation coefficient) r>0.80 and P<0.05. Finally, 197 and 113 differential metabolites and proteins were screened, respectively, in patients with CR/PR and SD/PD groups. The KEGG enrichment analysis revealed that all of these metabolites and proteins were enriched in cholesterol metabolism and in the NF-κB and phospholipase D signalling pathways. The present study is the first to demonstrate that PD-1 inhibitor resistance may be attributed to cholesterol metabolism or NF-κB and phospholipase D signalling pathway activation. This finding suggests that targeting these signalling pathways may be a promising novel therapeutic approach in OSCC which may improve prognosis in patients undergoing immunotherapy.

Keywords: PD-1 monoclonal antibody; immunotherapy; metabolics; oesophageal squamous cell carcinoma; proteomics.

Conflict of interest statement

The authors declare that they have no competing interests.

Copyright © 2020, Spandidos Publications.

Figures

Figure 1
Figure 1
Workflow of the present study. The plasmas of patients with oesophageal squamous cell carcinoma, who were treatment with anti-programmed death receptor-1 monoclonal antibody combined with chemotherapy after the fourth treatment cycle, were collected. The curative effect was simultaneously determined by the Responsive Evaluation Criteria in Solid Tumours 1.1 at the same period. Finally, the plasma samples were divided into a complete response/partial response group (n=15) and a stable disease/progressive disease group (n=16). The DEPs and DEMs between the two groups, which may mainly contribute to the immunotherapy resistance, were then identified by proteomic (data-independent acquisition proteomics) and metabolic (including TM-widely targeted metabolomics and widely targeted lipidomics) analysis. Furthermore, conjoint analysis of the DEPs and DEMs included Kyoto Encyclopedia of Genes and Genomes enrichment pathway analysis and correlated analysis was used to elucidate the molecular mechanisms. CR/PR, complete response/partial response; SD/PD, stable disease/progressive disease; DEPs, differentially expressed proteins; DEMs, differentially expressed metabolites; DIA, data-independent acquisition; KEGG, Kyoto Encyclopedia of Genes and Genomes.
Figure 2
Figure 2
Metabolic change analysis of stable disease/progressive disease vs. complete response/partial response using TM-widely targeted metabolomics. (A) The volcano plot shows the DEMs in the two groups. Each dot in the volcano map represents a metabolite, with the green dots representing downregulated differential metabolites, the red dots representing upregulated differential metabolites, and the grey dots representing detected but not significantly different metabolites. The x-coordinate represents the logarithmic value (log2FC) of the multiple of the relative content difference of a certain metabolite in the two groups of samples. The greater the absolute value of the x-coordinate is, the greater the relative content difference of the substance between the two groups of samples. Under VIP + FC (fold change) double screening conditions: The ordinate represents the VIP value, and the larger the ordinate value, the more significant the difference, and the more reliable the differentially expressed metabolites obtained by screening. (B) The top 10 up- and downregulated DEMs of the two groups. The horizontal coordinate represents the cumulative number of substances ordered according to the difference multiple from the smallest to the largest, and the vertical coordinate represents the pair value with the difference multiple as base 2. Each point represents a substance; the green points represent the top 10 downregulated substances, and the red points represent the top 10 upregulated substances. (C) The Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis of the DEMs. The rich factor is the ratio of the number of DEMs in the corresponding pathway to the total number of metabolites detected by the pathway. The higher the value, the greater the enrichment degree. The abscissa represents the rich factor corresponding to each pathway; the ordinate represents the pathway name; the colour of the dots is the P-value; the redder it is, the more significant the enrichment is. The size of the dot represents the number of enriched differential metabolites. DEMs, differentially expressed metabolites; VIP, variable importance in projection.
Figure 3
Figure 3
Metabolic change analysis of stable disease/progressive disease vs. complete response/partial response using widely targeted lipidomics. (A) The Volcano plot shows the DEMs in the two groups. Each dot in the volcano map represents a metabolite, with the green dots representing downregulated differential metabolites, the red dots representing upregulated differential metabolites, and the grey dots representing detected but not significantly different metabolites. The x-coordinate represents the logarithmic value (log2FC) of the multiple of the relative content difference of a certain metabolite in the two groups of samples. The greater the absolute value of the x-coordinate is, the greater the relative content difference of the substance between the two groups of samples. Under VIP + FC (fold change) double screening conditions: The ordinate represents the VIP value, and the larger the ordinate value, the more significant the difference, and the more reliable the DEMs obtained by screening. (B) The top10 up- and downregulated DEMs of the two groups. The horizontal coordinate represents the cumulative number of substances ordered according to the difference multiple from the smallest to the largest, and the vertical coordinate represents the pair value with the difference multiple as base 2. Each point represents a substance; the green points represent the top 10 downregulated substances, and the red points represent the top 10 upregulated substances. (C) The Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis of the DEMs. The rich factor is the ratio of the number of DEMs in the corresponding pathway to the total number of metabolites detected by the pathway. The higher the value, the greater the enrichment degree. The abscissa represents the rich factor corresponding to each pathway; the ordinate represents the pathway name; the colour of the dots is the P-value; the redder it is, the more significant the enrichment is. The size of the dot represents the number of enriched differential metabolites. DEMs, differentially expressed metabolites; VIP, variable importance in projection.
Figure 4
Figure 4
Protein profiling changes of stable disease/progressive disease vs. complete response/partial response using quantitative proteomics. (A) The volcano plot shows the DEPs in the two groups. The horizontal coordinate represents log2FC, the vertical coordinate represents-log10(P-value), the red and blue scatter plots represent the upregulated and downregulated proteins, respectively, and the dark grey scatter plots represent the non-significantly expressed proteins. (B) The heat map of the protein profiling of the two groups. The row represents the protein cluster, the column represents the sample cluster, and the shorter the cluster branch, the larger the similarity. (C) The top 20 GO biological processes of the DEPs. The x-coordinate represents the enrichment ratio (GeneRatio/BgRatio). The greater the enrichment ratio, the greater the degree of enrichment of the differential protein. The y-coordinate represents the enriched GO items. The change of dot colour from blue to red represents the change in the P-value from large to small. The smaller the P-value, the more statistically significant it is. The size of the dot represents the number of different proteins annotated by the corresponding item. (D) The top 20 KEGG pathways of the DEPs. The x-coordinate represents the enrichment ratio (GeneRatio/BgRatio). The larger the enrichment ratio is, the higher the enrichment degree of differential protein is. The y-coordinate represents the enriched KEGG pathway. The change of dot colour from blue to red represents the change of the P-value from large to small. The smaller the P-value is, the more statistically significant it is. The size of the dot represents the number of different proteins in the corresponding functional annotation. DEPs, differentially expressed proteins; GO, Gene Ontology; KEGG, Kyoto Encyclopedia of Genes and Genomes; SD-PD, stable disease-progressive disease; CR-PR, complete response-partial response.
Figure 5
Figure 5
Conjoint analyses of the metabolics and proteomics data. (A and B) The bubble diagram shows the co-enrichment KEGG pathways of the diferentially expressed metabolites and the differentially expressed proteins of the (A) widely targeted lipidomics and (B) TM-widely targeted metabolomics. The x-coordinate represents the enrichment factors (Diff/Background) of the pathway in different omics, and the y-coordinate represents the names of KEGG pathways. The gradient of red, yellow and green represents the change of enrichment significance from high to medium to low, which is represented by the P-value. The shape of the bubble represents different omics, and the size of the bubble represents the number of different metabolites or proteins, and the larger the number, the larger the dot. KEGG, Kyoto Encyclopedia of Genes and Genomes.
Figure 6
Figure 6
Enrichment of DEMs and DEPs in the cholesterol metabolism, NF-kappaB and phospholipase D signaling pathways. (A) Enrichment of DEMs and DEPs in the cholesterol metabolism signaling pathway. (B) Enrichment of DEMs and DEPs in the NF-kappaB signaling pathway. (C) Enrichment of DEMs and DEPs in the phospholipase D signaling pathway. (D) The correlation network of the DEMs and DEPs in the cholesterol metabolism signaling pathway. Blue circles represent metabolites, red circles represent proteins, red lines represent positive correlations, and blue lines represent negative correlations. (E) The correlation network of the DEMs and DEPs in the bile secretion signaling pathway. Blue circles represent metabolites, red circles represent proteins, red lines represent positive correlations, and blue lines represent negative correlations. (F) The cholesterol metabolism pathway; the upregulated metabolites are indicated in red. DEMs, differentially expressed metabolites; DEPs, differentially expressed proteins; SD-PD, stable disease-progressive disease; CR-PR, complete response-partial response; APOH, apolipoprotein H.

References

    1. Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of cancer: Clinical impact and mechanisms of response and resistance. Annu Rev Pathol. 2021;16:223–249. doi: 10.1146/annurev-pathol-042020-042741.
    1. Carlino MS, Larkin J, Long GV. Immune checkpoint inhibitors in melanoma. Lancet. 2021;398:1002–1014. doi: 10.1016/S0140-6736(21)01206-X.
    1. Li F, Chen Y, Pang M, Yang P, Jing H. Immune checkpoint inhibitors and cellular treatment for lymphoma immunotherapy. Clin Exp Immunol. 2021;205:1–11. doi: 10.1111/cei.13592.
    1. Kojima T, Shah MA, Muro K, Francois E, Adenis A, Hsu CH, Doi T, Moriwaki T, Kim SB, Lee SH, et al. Randomized phase III KEYNOTE-181 study of pembrolizumab versus chemotherapy in advanced esophageal cancer. J Clin Oncol. 2020;38:4138–4148. doi: 10.1200/JCO.20.01888.
    1. Kato K, Cho BC, Takahashi M, Okada M, Lin CY, Chin K, Kadowaki S, Ahn MJ, Hamamoto Y, Doki Y, et al. Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019;20:1506–1517. doi: 10.1016/S1470-2045(19)30626-6.
    1. Huang J, Xu J, Chen Y, Zhuang W, Zhang Y, Chen Z, Chen J, Zhang H, Niu Z, Fan Q, et al. Camrelizumab versus investigator's choice of chemotherapy as second-line therapy for advanced or metastatic oesophageal squamous cell carcinoma (ESCORT): A multicentre, randomised, open-label, phase 3 study. Lancet Oncol. 2020;21:832–842. doi: 10.1016/S1470-2045(20)30110-8.
    1. Sun JM, Shen L, Shah MA, Enzinger P, Adenis A, Doi T, Kojima T, Metges JP, Li Z, Kim SB, et al. Pembrolizumab plus chemotherapy versus chemotherapy alone for first-line treatment of advanced oesophageal cancer (KEYNOTE-590): A randomised, placebo-controlled, phase 3 study. Lancet. 2021;398:759–771. doi: 10.1016/S0140-6736(21)01234-4.
    1. Luo H, Lu J, Bai Y, Mao T, Wang J, Fan Q, Zhang Y, Zhao K, Chen Z, Gao S, et al. Effect of camrelizumab vs placebo added to chemotherapy on survival and progression-free survival in patients with advanced or metastatic esophageal squamous cell carcinoma: The ESCORT-1st randomized clinical trial. JAMA. 2021;326:916–925. doi: 10.1001/jama.2021.12836.
    1. Rinschen MM, Ivanisevic J, Giera M, Siuzdak G. Identification of bioactive metabolites using activity metabolomics. Nat Rev Mol Cell Biol. 2019;20:353–367. doi: 10.1038/s41580-019-0108-4.
    1. Muthubharathi BC, Gowripriya T, Balamurugan K. Metabolomics: Small molecules that matter more. Mol Omics. 2021;17:210–229. doi: 10.1039/d0mo00176g.
    1. Chen PH, Cai L, Huffman K, Yang C, Kim J, Faubert B, Boroughs L, Ko B, Sudderth J, McMillan EA, et al. Metabolic diversity in human non-small cell lung cancer cells. Mol Cell. 2019;76:838–851.e5. doi: 10.1016/j.molcel.2019.08.028.
    1. Shen Y, Sun M, Zhu J, Wei M, Li H, Zhao P, Wang J, Li R, Tian L, Tao Y, et al. Tissue metabolic profiling reveals major metabolic alteration in colorectal cancer. Mol Omics. 2021;17:464–471. doi: 10.1039/d1mo00022e.
    1. Li L, Ma J. Molecular characterization of metabolic subtypes of gastric cancer based on metabolism-related lncRNA. Sci Rep. 2021;11(21491) doi: 10.1038/s41598-021-00410-7.
    1. Chen CJ, Lee DY, Yu J, Lin YN, Lin TM. Recent advances in LC-MS-based metabolomics for clinical biomarker discovery. Mass Spectrom Rev. 2022;(e21785) doi: 10.1002/mas.21785. (Epub ahead of print)
    1. Seger C, Salzmann L. After another decade: LC-MS/MS became routine in clinical diagnostics. Clin Biochem. 2020;82:2–11. doi: 10.1016/j.clinbiochem.2020.03.004.
    1. Beccaria M, Cabooter D. Current developments in LC-MS for pharmaceutical analysis. Analyst. 2020;145:1129–1157. doi: 10.1039/c9an02145k.
    1. Zhang S, Wang H, Zhu MJ. A sensitive GC/MS detection method for analyzing microbial metabolites short chain fatty acids in fecal and serum samples. Talanta. 2019;196:249–254. doi: 10.1016/j.talanta.2018.12.049.
    1. Beale DJ, Pinu FR, Kouremenos KA, Poojary MM, Narayana VK, Boughton BA, Kanojia K, Dayalan S, Jones OAH, Dias DA. Review of recent developments in GC-MS approaches to metabolomics-based research. Metabolomics. 2018;14(152) doi: 10.1007/s11306-018-1449-2.
    1. Hu R, Li T, Yang Y, Tian Y, Zhang L. NMR-based metabolomics in cancer research. Adv Exp Med Biol. 2021;1280:201–218. doi: 10.1007/978-3-030-51652-9_14.
    1. Bichmann L, Gupta S, Rosenberger G, Kuchenbecker L, Sachsenberg T, Ewels P, Alka O, Pfeuffer J, Kohlbacher O, Röst H. DIAproteomics: A multifunctional data analysis pipeline for data-independent acquisition proteomics and peptidomics. J Proteome Res. 2021;20:3758–3766. doi: 10.1021/acs.jproteome.1c00123.
    1. Ma L, Muscat JE, Sinha R, Sun D, Xiu G. Proteomics of exhaled breath condensate in lung cancer and controls using data-independent acquisition (DIA): A pilot study. J Breath Res. 2021;15(026002) doi: 10.1088/1752-7163/abd07e.
    1. Krasny L, Huang PH. Data-independent acquisition mass spectrometry (DIA-MS) for proteomic applications in oncology. Mol Omics. 2021;17:29–42. doi: 10.1039/d0mo00072h.
    1. Zheng X, Xu K, Zhou B, Chen T, Huang Y, Li Q, Wen F, Ge W, Wang J, Yu S, et al. A circulating extracellular vesicles-based novel screening tool for colorectal cancer revealed by shotgun and data-independent acquisition mass spectrometry. J Extracell Vesicles. 2020;9(1750202) doi: 10.1080/20013078.2020.1750202.
    1. Rao J, Wan X, Tou F, He Q, Xiong A, Chen X, Cui W, Zheng Z. Molecular characterization of advanced colorectal cancer using serum proteomics and metabolomics. Front Mol Biosci. 2021;8(687229) doi: 10.3389/fmolb.2021.687229.
    1. Zhang Q, Zhang Y, Sun S, Wang K, Qian J, Cui Z, Tao T, Zhou J. ACOX2 is a prognostic marker and impedes the progression of hepatocellular carcinoma via PPARα pathway. Cell Death Dis. 2021;12(15) doi: 10.1038/s41419-020-03291-2.
    1. Kong R, Qian X, Ying W. Pancreatic cancer cells spectral library by DIA-MS and the phenotype analysis of gemcitabine sensitivity. Sci Data. 2022;9(283) doi: 10.1038/s41597-022-01407-1.
    1. Keam SP, Gulati T, Gamell C, Caramia F, Huang C, Schittenhelm RB, Kleifeld O, Neeson PJ, Haupt Y, Williams SG. Exploring the oncoproteomic response of human prostate cancer to therapeutic radiation using data-independent acquisition (DIA) mass spectrometry. Prostate. 2018;78:563–575. doi: 10.1002/pros.23500.
    1. Sun Y, Li L, Zhou Y, Ge W, Wang H, Wu R, Liu W, Chen H, Xiao Q, Cai X, et al. Stratification of follicular thyroid tumours using data-independent acquisition proteomics and a comprehensive thyroid tissue spectral library. Mol Oncol. 2022;16:1611–1624. doi: 10.1002/1878-0261.13198.
    1. Oken MM, Creech RH, Tormey DC, Horton J, Davis TE, McFadden ET, Carbone PP. Toxicity and response criteria of the Eastern cooperative oncology group. Am J Clin Oncol. 1982;5:649–655.
    1. Schwartz LH, Litière S, de Vries E, Ford R, Gwyther S, Mandrekar S, Shankar L, Bogaerts J, Chen A, Dancey J, et al. RECIST 1.1-update and clarification: From the RECIST committee. Eur J Cancer. 2016;62:132–137. doi: 10.1016/j.ejca.2016.03.081.
    1. Greene FL, Page DL, Fleming ID, Fritz AG, Balch CM, Haller DG, Morrow M (eds) AJCC cancer staging manual. 6th edition. American Joint Committe on Cancer, Chicago, IL, pp91-99, 2002.
    1. Demichev V, Messner CB, Vernardis SI, Lilley KS, Ralser M. DIA-NN: Neural networks and interference correction enable deep proteome coverage in high throughput. Nat Methods. 2020;17:41–44. doi: 10.1038/s41592-019-0638-x.
    1. Yu G, Wang LG, Han Y, He QY. clusterProfiler: An R package for comparing biological themes among gene clusters. OMICS. 2012;16:284–287. doi: 10.1089/omi.2011.0118.
    1. Yang T, Hui R, Nouws J, Sauler M, Zeng T, Wu Q. Untargeted metabolomics analysis of esophageal squamous cell cancer progression. J Transl Med. 2022;20(127) doi: 10.1186/s12967-022-03311-z.
    1. White L, Ma J, Liang S, Sanchez-Espiridion B, Liang D. LC-MS/MS determination of d-mannose in human serum as a potential cancer biomarker. J Pharm Biomed Anal. 2017;137:54–59. doi: 10.1016/j.jpba.2016.12.017.
    1. Mir SA, Rajagopalan P, Jain AP, Khan AA, Datta KK, Mohan SV, Lateef SS, Sahasrabuddhe N, Somani BL, Keshava Prasad TS, et al. LC-MS-based serum metabolomic analysis reveals dysregulation of phosphatidylcholines in esophageal squamous cell carcinoma. J Proteomics. 2015;127:96–102. doi: 10.1016/j.jprot.2015.05.013.
    1. Zhang J, Bowers J, Liu L, Wei S, Gowda GA, Hammoud Z, Raftery D. Esophageal cancer metabolite biomarkers detected by LC-MS and NMR methods. PLoS One. 2012;7(e30181) doi: 10.1371/journal.pone.0030181.
    1. Liang S, Sanchez-Espiridion B, Xie H, Ma J, Wu X, Liang D. Determination of proline in human serum by a robust LC-MS/MS method: Application to identification of human metabolites as candidate biomarkers for esophageal cancer early detection and risk stratification. Biomed Chromatogr. 2015;29:570–577. doi: 10.1002/bmc.3315.
    1. Song E, Zhu R, Hammoud ZT, Mechref Y. LC-MS/MS quantitation of esophagus disease blood serum glycoproteins by enrichment with hydrazide chemistry and lectin affinity chromatography. J Proteome Res. 2014;13:4808–4820. doi: 10.1021/pr500570m.
    1. Zhang H, Wang L, Hou Z, Ma H, Mamtimin B, Hasim A, Sheyhidin I. Metabolomic profiling reveals potential biomarkers in esophageal cancer progression using liquid chromatography-mass spectrometry platform. Biochem Biophys Res Commun. 2017;491:119–125. doi: 10.1016/j.bbrc.2017.07.060.
    1. Guo JH, Xing GL, Fang XH, Wu HF, Zhang B, Yu JZ, Fan ZM, Wang LD. Proteomic profiling of fetal esophageal epithelium, esophageal cancer, and tumor-adjacent esophageal epithelium and immunohistochemical characterization of a representative differential protein, PRX6. World J Gastroenterol. 2017;23:1434–1442. doi: 10.3748/wjg.v23.i8.1434.
    1. Yazdian-Robati R, Ahmadi H, Riahi MM, Lari P, Aledavood SA, Rashedinia M, Abnous K, Ramezani M. Comparative proteome analysis of human esophageal cancer and adjacent normal tissues. Iran J Basic Med Sci. 2017;20:265–271. doi: 10.22038/ijbms.2017.8354.
    1. O'Neill JR, Pak HS, Pairo-Castineira E, Save V, Paterson-Brown S, Nenutil R, Vojtěšek B, Overton I, Scherl A, Hupp TR. Quantitative shotgun proteomics unveils candidate novel esophageal adenocarcinoma (EAC)-specific proteins. Mol Cell Proteomics. 2017;16:1138–1150. doi: 10.1074/mcp.M116.065078.
    1. Schwacke J, Millar TP, Hammond CE, Saha A, Hoffman BJ, Romagnuolo J, Hill EG, Smolka AJ. Discrimination of normal and esophageal cancer plasma proteomes by MALDI-TOF mass spectrometry. Dig Dis Sci. 2015;60:1645–1654. doi: 10.1007/s10620-014-3513-8.
    1. Wang D, Chen J, Han J, Wang K, Fang W, Jin J, Xue S. iTRAQ and two-dimensional-LC-MS/MS reveal NAA10 is a potential biomarker in esophageal squamous cell carcinoma. Proteomics Clin Appl. 2022;16(e2100081) doi: 10.1002/prca.202100081.
    1. Singh S, Bano A, Saraya A, Das P, Sharma R. iTRAQ-based analysis for the identification of MARCH8 targets in human esophageal squamous cell carcinoma. J Proteomics. 2021;236(104125) doi: 10.1016/j.jprot.2021.104125.
    1. Wang X, Peng Y, Xie M, Gao Z, Yin L, Pu Y, Liu R. Identification of extracellular matrix protein 1 as a potential plasma biomarker of ESCC by proteomic analysis using iTRAQ and 2D-LC-MS/MS. Proteomics Clin Appl. 2017;11(1600163) doi: 10.1002/prca.201600163.
    1. Deng F, Zhou K, Li Q, Liu D, Li M, Wang H, Zhang W, Ma Y. iTRAQ-based quantitative proteomic analysis of esophageal squamous cell carcinoma. Tumour Biol. 2016;37:1909–1918. doi: 10.1007/s13277-015-3840-1.
    1. Liu W, Xie L, He YH, Wu ZY, Liu LX, Bai XF, Deng DX, Xu XE, Liao LD, Lin W, et al. Large-scale and high-resolution mass spectrometry-based proteomics profiling defines molecular subtypes of esophageal cancer for therapeutic targeting. Nat Commun. 2021;12(4961) doi: 10.1038/s41467-021-25202-5.
    1. Li Y, Yang B, Ma Y, Peng X, Wang Z, Sheng B, Wei Z, Cui Y, Liu Z. Phosphoproteomics reveals therapeutic targets of esophageal squamous cell carcinoma. Signal Transduct Target Ther. 2021;6(381) doi: 10.1038/s41392-021-00682-5.
    1. Joyce D, Albanese C, Steer J, Fu M, Bouzahzah B, Pestell RG. NF-kappaB and cell-cycle regulation: the cyclin connection. Cytokine Growth Factor Rev. 2001;12:73–90. doi: 10.1016/s1359-6101(00)00018-6.
    1. Grivennikov SI, Karin M. Dangerous liaisons: STAT3 and NF-kappaB collaboration and crosstalk in cancer. Cytokine Growth Factor Rev. 2010;21:11–19. doi: 10.1016/j.cytogfr.2009.11.005.
    1. Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H, Vandenabeele P. Regulated necrosis: The expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol. 2014;15:135–147. doi: 10.1038/nrm3737.
    1. Taniguchi K, Wu LW, Grivennikov SI, de Jong PR, Lian I, Yu FX, Wang K, Ho SB, Boland BS, Chang JT, et al. A gp130-Src-YAP module links inflammation to epithelial regeneration. Nature. 2015;519:57–62. doi: 10.1038/nature14228.
    1. Johnson RF, Perkins ND. Nuclear factor-κB, p53, and mitochondria: Regulation of cellular metabolism and the Warburg effect. Trends Biochem Sci. 2012;37:317–324. doi: 10.1016/j.tibs.2012.04.002.
    1. Xia Y, Shen S, Verma IM. NF-κB, an active player in human cancers. Cancer Immunol Res. 2014;2:823–830. doi: 10.1158/2326-6066.CIR-14-0112.
    1. Wu Y, Zhou BP. Inflammation: A driving force speeds cancer metastasis. Cell Cycle. 2009;8:3267–3273. doi: 10.4161/cc.8.20.9699.
    1. Amato CM, Hintzsche JD, Wells K, Applegate A, Gorden NT, Vorwald VM, Tobin RP, Nassar K, Shellman YG, Kim J, et al. Pre-treatment mutational and transcriptomic landscape of responding metastatic melanoma patients to anti-PD1 immunotherapy. Cancers (Basel) 2020;12(1943) doi: 10.3390/cancers12071943.
    1. Roh W, Chen PL, Reuben A, Spencer CN, Prieto PA, Miller JP, Gopalakrishnan V, Wang F, Cooper ZA, Reddy SM, et al. Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance. Sci Transl Med. 2017;9(eaah3560) doi: 10.1126/scitranslmed.aah3560.
    1. Shi LZ, Fu T, Guan B, Chen J, Blando JM, Allison JP, Xiong L, Subudhi SK, Gao J, Sharma P. Interdependent IL-7 and IFN-γ signalling in T-cell controls tumour eradication by combined α-CTLA-4+α-PD-1 therapy. Nat Commun. 2016;7(12335) doi: 10.1038/ncomms12335.
    1. Dong MB, Wang G, Chow RD, Ye L, Zhu L, Dai X, Park JJ, Kim HR, Errami Y, Guzman CD, et al. Systematic immunotherapy target discovery using genome-scale in vivo CRISPR screens in CD8 T cells. Cell. 2019;178:1189–1204.e23. doi: 10.1016/j.cell.2019.07.044.
    1. Li G, Boucher JC, Kotani H, Park K, Zhang Y, Shrestha B, Wang X, Guan L, Beatty N, Abate-Daga D, Davila ML. 4-1BB enhancement of CAR T function requires NF-κB and TRAFs. JCI Insight. 2018;3(e121322) doi: 10.1172/jci.insight.121322.
    1. Philipson BI, O'Connor RS, May MJ, June CH, Albelda SM, Milone MC. 4-1BB costimulation promotes CAR T cell survival through noncanonical NF-κB signaling. Sci Signal. 2020;13(eaay8248) doi: 10.1126/scisignal.aay8248.
    1. Schumacher LY, Vo DD, Garban HJ, Comin-Anduix B, Owens SK, Dissette VB, Glaspy JA, McBride WH, Bonavida B, Economou JS, Ribas A. Immunosensitization of tumor cells to dendritic cell-activated immune responses with the proteasome inhibitor bortezomib (PS-341, Velcade) J Immunol. 2006;176:4757–4765. doi: 10.4049/jimmunol.176.8.4757.
    1. Enzler T, Sano Y, Choo MK, Cottam HB, Karin M, Tsao H, Park JM. Cell-selective inhibition of NF-κB signaling improves therapeutic index in a melanoma chemotherapy model. Cancer Discov. 2011;1:496–507. doi: 10.1158/-11-0143.
    1. Xiao Z, Su Z, Han S, Huang J, Lin L, Shuai X. Dual pH-sensitive nanodrug blocks PD-1 immune checkpoint and uses T cells to deliver NF-κB inhibitor for antitumor immunotherapy. Sci Adv. 2020;6(eaay7785) doi: 10.1126/sciadv.aay7785.
    1. Cai M, He J, Xiong J, Tay LW, Wang Z, Rog C, Wang J, Xie Y, Wang G, Banno Y, et al. Phospholipase D1-regulated autophagy supplies free fatty acids to counter nutrient stress in cancer cells. Cell Death Dis. 2016;7(e2448) doi: 10.1038/cddis.2016.355.
    1. Palikaras K, Lionaki E, Tavernarakis N. Mechanisms of mitophagy in cellular homeostasis, physiology and pathology. Nat Cell Biol. 2018;20:1013–1022. doi: 10.1038/s41556-018-0176-2.
    1. Ganesan R, Mallets E, Gomez-Cambronero J. The transcription factors Slug (SNAI2) and Snail (SNAI1) regulate phospholipase D (PLD) promoter in opposite ways towards cancer cell invasion. Mol Oncol. 2016;10:663–676. doi: 10.1016/j.molonc.2015.12.006.
    1. Ho HY, Lin CW, Chien MH, Reiter RJ, Su SC, Hsieh YH, Yang SF. Melatonin suppresses TPA-induced metastasis by downregulating matrix metalloproteinase-9 expression through JNK/SP-1 signaling in nasopharyngeal carcinoma. J Pineal Res. 2016;61:479–492. doi: 10.1111/jpi.12365.
    1. Issuree PD, Pushparaj PN, Pervaiz S, Melendez AJ. Resveratrol attenuates C5a-induced inflammatory responses in vitro and in vivo by inhibiting phospholipase D and sphingosine kinase activities. FASEB J. 2009;23:2412–2424. doi: 10.1096/fj.09-130542.
    1. Park MH, Min do S. Quercetin-induced downregulation of phospholipase D1 inhibits proliferation and invasion in U87 glioma cells. Biochem Biophys Res Commun. 2011;412:710–715. doi: 10.1016/j.bbrc.2011.08.037.
    1. Garcia A, Zheng Y, Zhao C, Toschi A, Fan J, Shraibman N, Brown HA, Bar-Sagi D, Foster DA, Arbiser JL. Honokiol suppresses survival signals mediated by Ras-dependent phospholipase D activity in human cancer cells. Clin Cancer Res. 2008;14:4267–4274. doi: 10.1158/1078-0432.CCR-08-0102.
    1. Huang B, Song BL, Xu C. Cholesterol metabolism in cancer: Mechanisms and therapeutic opportunities. Nat Metab. 2020;2:132–141. doi: 10.1038/s42255-020-0174-0.
    1. Voisin M, de Medina P, Mallinger A, Dalenc F, Huc-Claustre E, Leignadier J, Serhan N, Soules R, Ségala G, Mougel A, et al. Identification of a tumor-promoter cholesterol metabolite in human breast cancers acting through the glucocorticoid receptor. Proc Natl Acad Sci USA. 2017;114:E9346–E9355. doi: 10.1073/pnas.1707965114.
    1. Chimento A, Casaburi I, Avena P, Trotta F, De Luca A, Rago V, Pezzi V, Sirianni R. Cholesterol and its metabolites in tumor growth: Therapeutic potential of statins in cancer treatment. Front Endocrinol (Lausanne) 2019;9(807) doi: 10.3389/fendo.2018.00807.
    1. Ding X, Zhang W, Li S, Yang H. The role of cholesterol metabolism in cancer. Am J Cancer Res. 2019;9:219–227.
    1. Wang Y, Liu C, Hu L. Cholesterol regulates cell proliferation and apoptosis of colorectal cancer by modulating miR-33a-PIM3 pathway. Biochem Biophys Res Commun. 2019;511:685–692. doi: 10.1016/j.bbrc.2019.02.123.
    1. Liu Z, Liu X, Liu S, Cao Q. Cholesterol promotes the migration and invasion of renal carcinoma cells by regulating the KLF5/miR-27a/FBXW7 pathway. Biochem Biophys Res Commun. 2018;502:69–75. doi: 10.1016/j.bbrc.2018.05.122.
    1. Raccosta L, Fontana R, Maggioni D, Lanterna C, Villablanca EJ, Paniccia A, Musumeci A, Chiricozzi E, Trincavelli ML, Daniele S, et al. The oxysterol-CXCR2 axis plays a key role in the recruitment of tumor-promoting neutrophils. J Exp Med. 2013;210:1711–1728. doi: 10.1084/jem.20130440.
    1. Condamine T, Dominguez GA, Youn JI, Kossenkov AV, Mony S, Alicea-Torres K, Tcyganov E, Hashimoto A, Nefedova Y, Lin C, et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci Immunol. 2016;1(aaf8943) doi: 10.1126/sciimmunol.aaf8943.
    1. Goossens P, Rodriguez-Vita J, Etzerodt A, Masse M, Rastoin O, Gouirand V, Ulas T, Papantonopoulou O, Van Eck M, Auphan-Anezin N, et al. Membrane cholesterol efflux drives tumor-associated macrophage reprogramming and tumor progression. Cell Metab. 2019;29:1376–1389.e4. doi: 10.1016/j.cmet.2019.02.016.
    1. Kidani Y, Elsaesser H, Hock MB, Vergnes L, Williams KJ, Argus JP, Marbois BN, Komisopoulou E, Wilson EB, Osborne TF, et al. Sterol regulatory element-binding proteins are essential for the metabolic programming of effector T cells and adaptive immunity. Nat Immunol. 2013;14:489–499. doi: 10.1038/ni.2570.
    1. Larsen SB, Dehlendorff C, Skriver C, Dalton SO, Jespersen CG, Borre M, Brasso K, Nørgaard M, Johansen C, Sørensen HT, et al. Postdiagnosis statin use and mortality in danish patients with prostate cancer. J Clin Oncol. 2017;35:3290–3297. doi: 10.1200/JCO.2016.71.8981.
    1. Xia Y, Xie Y, Yu Z, Xiao H, Jiang G, Zhou X, Yang Y, Li X, Zhao M, Li L, et al. The mevalonate pathway is a druggable target for vaccine adjuvant discovery. Cell. 2018;175:1059–1073.e21. doi: 10.1016/j.cell.2018.08.070.
    1. Maione F, Oliaro-Bosso S, Meda C, Di Nicolantonio F, Bussolino F, Balliano G, Viola F, Giraudo E. The cholesterol biosynthesis enzyme oxidosqualene cyclase is a new target to impair tumour angiogenesis and metastasis dissemination. Sci Rep. 2015;5(9054) doi: 10.1038/srep09054.
    1. Lanterna C, Musumeci A, Raccosta L, Corna G, Moresco M, Maggioni D, Fontana R, Doglioni C, Bordignon C, Traversari C, Russo V. The administration of drugs inhibiting cholesterol/oxysterol synthesis is safe and increases the efficacy of immunotherapeutic regimens in tumor-bearing mice. Cancer Immunol Immunother. 2016;65:1303–1315. doi: 10.1007/s00262-016-1884-8.
    1. Lu Y, Wang Y, Qiu Y, Xuan W. Analysis of the relationship between the expression level of TTR and APOH and prognosis in patients with colorectal cancer metastasis based on bioinformatics. Contrast Media Mol Imaging. 2022;2022(1121312) doi: 10.1155/2022/1121312.
    1. Li X, Wang L, Wang L, Feng Z, Peng C. Single-cell sequencing of hepatocellular carcinoma reveals cell interactions and cell heterogeneity in the microenvironment. Int J Gen Med. 2021;14:10141–10153. doi: 10.2147/IJGM.S338090.
    1. Mandili G, Notarpietro A, Khadjavi A, Allasia M, Battaglia A, Lucatello B, Frea B, Turrini F, Novelli F, Giribaldi G, Destefanis P. Beta-2-glycoprotein-1 and alpha-1-antitrypsin as urinary markers of renal cancer in von Hippel-Lindau patients. Biomarkers. 2018;23:123–130. doi: 10.1080/1354750X.2016.1269132.
    1. Fan T, Lu Z, Liu Y, Wang L, Tian H, Zheng Y, Zheng B, Xue L, Tan F, Xue Q, et al. A novel immune-related seventeen-gene signature for predicting early stage lung squamous cell carcinoma prognosis. Front Immunol. 2021;12(665407) doi: 10.3389/fimmu.2021.665407.
    1. Qiu XT, Song YC, Liu J, Wang ZM, Niu X, He J. Identification of an immune-related gene-based signature to predict prognosis of patients with gastric cancer. World J Gastrointest Oncol. 2020;12:857–876. doi: 10.4251/wjgo.v12.i8.857.

Source: PubMed

3
Předplatit