Welcome to visit Zhongnan Medical Journal Press Series journal website!

Home Articles Vol 38,2025 No.12 Detail

Analysis of key genes for gemcitabine resistance and their pan-cancer expression, immune infiltration and survival associations

Published on Dec. 31, 2025Total Views: 48 times Total Downloads: 8 times Download Mobile

Author: SHEN Ruibo 1 YANG Yulin 2 ZHANG Xin 2 LIANG Xinyi 2 YANG Huan 2 DU Juan 3

Affiliation: 1. First School of Clinical Medical, Gansu University of Chinese Medicine, Lanzhou 730000, China 2. Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China 3. School of Nursing, Gansu University of Chinese Medicine, Lanzhou 730000, China

Keywords: Gemcitabine Drug resistance Key genes Pan-cancer Immune infiltration Survival associations

DOI: 10.12173/j.issn.1004-4337.202504096

Reference: Shen RB, Yang YL, Zhang X, Liang XY, Yang H, Du J. Analysis of key genes for gemcitabine resistance and their pan-cancer expression, immune infiltration and survival associations[J]. Journal of Mathematical Medicine, 2025, 38(12): 936-948. DOI: 10.12173/j.issn.1004-4337.202504096[Article in Chinese]

  • Abstract
  • Full-text
  • References
Abstract

Objective  To explore the pan-cancer expression, immune infiltration, survival associations and drug sensitivity of key gemcitabine resistance genes, and to seek new alternative clinical therapies.

Methods  The gemcitabine resistance dataset was obtained through the GEO database. Differently expressed genes were screened, and the core differentially expressed genes were obtained through integrated analysis. The expression, immune infiltration, survival associations and drug sensitivity of key genes in pan-cancer were analyzed based on the cProSite, TIMER 2.0 and GSCA platforms.

Results  The key differential genes for gemcitabine resistance were SERPINB2 and CALB1. The expression of SERPINB2 was significantly increased in colon adenocarcinoma, lung squamous cell carcinoma, and skin cutaneous melanoma (P<0.001), and significantly decreased in breast invasive carcinoma, head and neck squamous cell carcinoma, kidney chromophobe, liver hepatocellular carcinoma, and lung adenocarcinoma (P<0.001). The expression of CALB1 was significantly increased in head and neck squamous cell carcinoma, lung squamous cell carcinoma and uterine corpus endometrial carcinoma (P<0.001), and significantly decreased in breast invasive carcinoma, kidney chromophobe, kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma and thyroid carcinoma (P<0.001). The expressions of SERPINB2 and CALB1 were negatively correlated with the infiltration levels of B cell, Gamma_delta T cell, CD4_T, etc., in various cancers; and were positively correlated with the infiltration levels of iTreg, nTreg, macrophage, etc. Compared to CALB1, the differential expression of SERPINB2 had a greater impact on the survival of cancer patients in pan-cancer. The correlation analysis results between gene expression and drug sensitivity showed that SERPINB2 was significantly positively correlated with belinostat (r=0.186, FDR<0.001). CALB1 had the highest positive correlation with BRD-K99006945 (r=0.137, FDR<0.05), and the highest negative correlation with PD 153035 (r=-0.246, FDR<0.001).

Conclusion  The abnormal expressions of key gemcitabine resistance genes SERPINB2 and CALB1 were closely related to the occurrence and development of various cancers. The expressions of SERPINB2 and CALB1 were associated with the poor prognosis of cancer patients. The expression levels of small molecules belinostat and BRD-K99006945 were significantly positively correlated with the key genes of gemcitabine resistance, and they might be candidate drugs with antagonistic potential, which required further research.

Full-text
Please download the PDF version to read the full text: download
References

1.Pandit B, Royzen M. Recent development of prodrugs of gemcitabine[J]. Genes(Basel), 2022, 13(3): 466. DOI: 10.3390/genes13030466.

2.Miao H, Chen X, Luan Y. Small molecular gemcitabine prodrugs for cancer therapy[J]. Curr Med Chem, 2020, 27(33): 5562-5582. DOI: 10.2174/0929867326666190816230650.

3.van der Heijden MS, Sonpavde G, Powles T, et al. Nivolumab plus gemcitabine-cisplatin in advanced urothelial carcinoma[J]. N Engl J Med, 2023, 389(19): 1778-1789. DOI: 10.1056/NEJMoa2309863.

4.McElree IM, Steinberg RL, Mott SL, et al. Comparison of sequential intravesical gemcitabine and docetaxel vs bacillus calmette-guérin for the treatment of patients with high-risk non-muscle-invasive bladder cancer[J]. JAMA Netw Open, 2023, 6(2): e230849. DOI: 10.1001/jamanetworkopen.2023.0849.

5.Coleman JA, Yip W, Wong NC, et al. Multicenter phase II clinical trial of gemcitabine and cisplatin as neoadjuvant chemotherapy for patients with high-grade upper tract urothelial carcinoma[J]. J Clin Oncol, 2023, 41(8): 1618-1625. DOI: 10.1200/JCO.22.00763.

6.Kim MJ, Kim HS, Kang HW, et al. SLC38A5 modulates ferroptosis to overcome gemcitabine resistance in pancreatic cancer[J]. Cells, 2023, 12(20): 2509. DOI: 10.3390/cells12202509.

7.Natu J, Nagaraju GP. Gemcitabine effects on tumor microenvironment of pancreatic ductal adenocarcinoma: special focus on resistance mechanisms and metronomic therapies[J]. Cancer Lett., 2023, 573: 216382. DOI: 10.1016/j.canlet.2023.216382.

8.Cordani M, Dando I, Ambrosini G, et al. Signaling, cancer cell plasticity, and intratumor heterogeneity[J]. Cell Commun Signal, 2024, 22(1): 255. DOI: 10.1186/s12964-024-01643-5.

9.Clough E, Barrett T, Wilhite SE, et al. NCBI GEO: archive for gene expression and epigenomics data sets: 23-year update[J]. Nucleic Acids Res, 2024, 52(D1): D138-D144. DOI: 10.1093/nar/gkad965.

10.Bardou P, Mariette J, Escudié F, et al. jvenn: an interactive Venn diagram viewer[J]. BMC Bioinformatics, 2014,15(1): 293. DOI: 10.1186/1471-2105-15-293.

11.Wang D, Qian X, Du YN, et al. cProSite: a web based interactive platform for online proteomics, phosphoproteomics, and genomics data analysis[J]. J Biotechnol Biomed, 2023, 6(4): 573-578. DOI: 10.26502/jbb.2642-91280119.

12.Li T, Fu J, Zeng Z, et al. TIMER2.0 for analysis of tumor-infiltrating immune cells[J]. Nucleic Acids Res, 2020, 48(W1): W509-W514. DOI: 10.1093/nar/gkaa407.

13.Liu CJ, Hu FF, Xie GY, et al. GSCA: an integrated platform for gene set cancer analysis at genomic, pharmacogenomic and immunogenomic levels[J]. Brief Bioinform, 2023, 24(1): bbac558. DOI: 10.1093/bib/bbac558.

14.Landau J, Tsaban L, Yaacov A, et al. Shared cancer dataset analysis identifies and predicts the quantitative effects of pan-cancer somatic driver variants[J]. Cancer Res, 2023, 83(1): 74-88. DOI: 10.1158/0008-5472.CAN-22-1038.

15.Kim S, Chen J, Cheng T, et al. PubChem 2023 update[J]. Nucleic Acids Res, 2023, 51(D1): D1373-D1380. DOI: 10.1093/nar/gkac956.

16.Rose AS, Bradley AR, Valasatava Y, et al. NGL viewer: web-based molecular graphics for large complexes[J]. Bioinformatics, 2018, 34(21): 3755-3758. DOI: 10.1093/bioinformatics/bty419.

17.Varamo C, Peraldo-Neia C, Ostano P, et al. Establishment and characterization of a new intrahepatic cholangiocarcinoma cell line resistant to gemcitabine[J]. Cancers(Basel), 2019, 11(4): 519. DOI: 10.3390/cancers11040519.

18.Nakano T, Saiki Y, Kudo C, et al. Acquisition of chemoresistance to gemcitabine is induced by a loss-of-function missense mutation of DCK[J]. Biochem Biophys Res Commun, 2015, 464(4): 1084-1089. DOI: 10.1016/j.bbrc.2015.07.080.

19.Zhou J, Zhang L, Zheng H, et al. Identification of chemoresistance-related mRNAs based on gemcitabine-resistant pancreatic cancer cell lines[J]. Cancer Med, 2020, 9(3): 1115-1130. DOI: 10.1002/cam4.2764.

20.Jankova L, Harrop SJ, Saunders DN, et al. Crystal structure of the complex of plasminogen activator inhibitor 2 with a peptide mimicking the reactive center loop[J]. J Biol Chem, 2001, 276(46): 43374-43382. DOI: 10.1074/jbc.M103021200.

21.Noble JW, Almalki R, Roe SM, et al. The X-ray structure of human calbindin-D28K: an improved model[J]. Acta Crystallogr D Struct Biol, 2018, 74(Pt 10): 1008-1014. DOI: 10.1107/S2059798318011610.

22.Sharpless NE, Sherr CJ. Forging a signature of in vivo senescence[J]. Nat Rev Cancer, 2015, 15(7): 397-408. DOI: 10.1038/nrc3960.

23.Hsieh HH, Chen YC, Jhan JR, et al. The serine protease inhibitor serpinB2 binds and stabilizes p21 in senescent cells[J]. J Cell Sci, 2017, 130(19): 3272-3281. DOI: 10.1242/jcs.204974.

24.Raynard C, Tessier N, Huna A, et al. Expression of the calcium-binding protein CALB1 is induced and controls intracellular Ca2+ levels in senescent cells[J]. Int J Mol Sci, 2022, 23(16): 9376. DOI: 10.3390/ijms23169376.

25.Cao LQ, Wang YN, Liang M, et al. CALB1 enhances the interaction between p53 and MDM2, and inhibits the senescence of ovarian cancer cells[J]. Mol Med Rep, 2019, 19(6): 5097-5104. DOI: 10.3892/mmr.2019.10212.

26.Westrick RJ, Røjkjaer LP, Yang AY, et al. Deficiency of plasminogen activator inhibitor-2 results in accelerated tumor growth[J]. J Thromb Haemost, 2020, 18(11): 2968-2975. DOI: 10.1111/jth.15054.

27.Kumar AA, Buckley BJ, Ranson M. The urokinase plasminogen activation system in pancreatic cancer: prospective diagnostic and therapeutic targets[J]. Biomolecules, 2022, 12(2): 152. DOI: 10.3390/biom12020152.

28.Furuya H, Hayashi K, Shimizu Y, et al. Plasminogen activator inhibitor-2 (PAI-2) overexpression supports bladder cancer development in PAI-1 knockout mice in N-butyl-N-(4-hydroxybutyl)-nitrosamine- induced bladder cancer mouse model[J]. J Transl Med, 2020, 18(1): 57. DOI: 10.1186/s12967-020-02239-6.

29.Qi L, Gao C, Feng F, et al. MicroRNAs associated with lung squamous cell carcinoma: New prognostic biomarkers and therapeutic targets[J]. J Cell Biochem, 2019, 120(11): 18956-18966. DOI: 10.1002/jcb.29216.

30.Yang W, Lu S, Peng L, et al. Integrated analysis of necroptosis-related genes for evaluating immune infiltration and colon cancer prognosis[J]. Front Immunol, 2022, 13: 1085038. DOI: 10.3389/fimmu.2022.1085038.

31.Piao YJ, Kim HS, Kim H, et al. SerpinB2 deficiency is associated with delayed mammary tumor development and decreased pro-tumorigenic macrophage polarization[J]. BMC Cancer, 2024, 24(1): 792. DOI: 10.1186/s12885-024-12473-6.

32.Jin T, Suk Kim H, Ki Choi S, et al. microRNA-200c/141 upregulates SerpinB2 to promote breast cancer cell metastasis and reduce patient survival[J]. Oncotarget, 2017, 8(20): 32769-32782. DOI: 10.18632/oncotarget.15680.

33.Piao YJ, Kim HS, Han W, et al. Transcriptome analysis of SerpinB2-deficient breast tumors provides insight into deciphering SerpinB2-mediated roles in breast cancer progression[J]. BMC Genomics, 2022, 23(1): 479. DOI: 10.1186/s12864-022-08704-4.

34.Cirillo F, Spinelli A, Talia M, et al. Estetrol/GPER/SERPINB2 transduction signaling inhibits the motility of triple-negative breast cancer cells[J]. J Transl Med, 2024, 22(1): 450. DOI: 10.1186/s12967-024-05269-6.

35.Shang S, Yang C, Chen F, et al. ID1 expressing macrophages support cancer cell stemness and limit CD8+ T cell infiltration in colorectal cancer[J]. Nat Commun, 2023, 14(1): 7661. DOI: 10.1038/s41467-023-43548-w.

36.Singh M, Kondraskhina AM, Hurst LD, et al. Staring at the onco-exaptation: the two-faced medley of an ancient retrovirus, HERVH[J]. J Clin Invest, 2023, 133(14): e172278. DOI: 10.1172/JCI172278.

37.El Omari N, Bakrim S, Khalid A, et al. Anticancer clinical efficiency and stochastic mechanisms of belinostat[J]. Biomed Pharmacother, 2023, 165: 115212. DOI: 10.1016/j.biopha.2023.115212.

38.Tollefsbol T. Handbook of epigenetics (3rd edition)[M]. Cambridge: Academic Press, 2023: 821-840.

39.Thorel L, Morice PM, Paysant H, et al. Comparative analysis of response to treatments and molecular features of tumor-derived organoids versus cell lines and PDX derived from the same ovarian clear cell carcinoma[J]. J Exp Clin Cancer Res, 2023, 42(1): 260. DOI: 10.1186/s13046-023-02809-8.

40.Ronellenfitsch MW, Zeiner PS, Mittelbronn M, et al. Akt and mTORC1 signaling as predictive biomarkers for the EGFR antibody nimotuzumab in glioblastoma[J]. Acta Neuropathol Commun, 2018, 6(1): 81. DOI: 10.1186/s40478-018-0583-4.

41.Mehus AA, Bergum N, Knutson P, et al. Activation of PPARγ and inhibition of cell proliferation reduces key proteins associated with the basal subtype of bladder cancer in As3+-transformed UROtsa cells[J]. PLoS One, 2020, 15(8): e0237976. DOI: 10.1371/journal.pone.0237976.

Popular papers
Last 6 months