Generic placeholder image

Current Bioinformatics

Editor-in-Chief

ISSN (Print): 1574-8936
ISSN (Online): 2212-392X

Research Article

A Pan-cancer Analysis Reveals the Tissue Specificity and Prognostic Impact of Angiogenesis-associated Genes in Human Cancers

Author(s): Zhenshen Bao, Minzhen Liao, Wanqi Dong, Yanhao Huo, Xianbin Li*, Peng Xu* and Wenbin Liu*

Volume 18, Issue 8, 2023

Published on: 07 July, 2023

Page: [670 - 679] Pages: 10

DOI: 10.2174/1574893618666230518163353

Price: $65

conference banner
Abstract

Introduction: Angiogenesis is one of the hallmarks of cancer and can impact the processes of cancer initiation, progression, and response to therapy.

Background: Anti-angiogenic therapy is thus an encouraging therapeutic option to treat cancers, but the detailed angiogenic mechanisms and the association between angiogenesis and clinical outcome remain unknown in different cancers.

Methods: Here, we systematically assess the impacts of 82 angiogenesis-associated genes (AAGs) in tumor tissue specificity and prognosis across 16 cancer types.

Results: Results demonstrate that the expression patterns of the 82 AAGs can reflect the tumor tissue specificity, and high expressions of up-regulated AAGs are significantly associated with poor prognosis of cancer. We further define a prognostic score for predicting overall survival (OS) based on the expressions of up-regulated AAGs and confirm its reliable predictive ability. Results indicate that a low prognostic score demonstrates a superior OS and vice versa.

Conclusion: The results of this study will contribute to the understanding of different tumor angiogenesis mechanisms in various tissues and cancer-personalized anti-angiogenic treatment. The code of our analysis can be accessed at https://github.com/ZhenshenBao/AAGs_analysis.git.

Graphical Abstract

[1]
Folkman J. Tumor angiogenesis. Adv Cancer Res 1985; 43: 175-203.
[http://dx.doi.org/10.1016/S0065-230X(08)60946-X] [PMID: 2581424]
[2]
Donnem T, Reynolds AR, Kuczynski EA, et al. Non-angiogenic tumours and their influence on cancer biology. Nat Rev Cancer 2018; 18(5): 323-36.
[http://dx.doi.org/10.1038/nrc.2018.14] [PMID: 29520090]
[3]
Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell 1996; 86(3): 353-64.
[http://dx.doi.org/10.1016/S0092-8674(00)80108-7] [PMID: 8756718]
[4]
Sherwood LM, Parris EE, Folkman J. Tumor angiogenesis: Therapeutic implications. N Engl J Med 1971; 285(21): 1182-6.
[http://dx.doi.org/10.1056/NEJM197111182852108] [PMID: 4938153]
[5]
Pepper MS, Montesano R, Mandriota SJ, Orci L, Vassalli JD. Angiogenesis: A paradigm for balanced extracellular proteolysis during cell migration and morphogenesis. Enzyme Protein 1996; 49(1-3): 138-62.
[http://dx.doi.org/10.1159/000468622] [PMID: 8797003]
[6]
Folkman J, Shing Y. Angiogenesis. J Biol Chem 1992; 267(16): 10931-4.
[http://dx.doi.org/10.1016/S0021-9258(19)49853-0] [PMID: 1375931]
[7]
Ramjiawan RR, Griffioen AW, Duda DG. Anti-angiogenesis for cancer revisited: Is there a role for combinations with immunotherapy? Angiogenesis 2017; 20(2): 185-204.
[http://dx.doi.org/10.1007/s10456-017-9552-y] [PMID: 28361267]
[8]
Yu WD, Sun G, Li J, Xu J, Wang X. Mechanisms and therapeutic potentials of cancer immunotherapy in combination with radiotherapy and/or chemotherapy. Cancer Lett 2019; 452: 66-70.
[http://dx.doi.org/10.1016/j.canlet.2019.02.048] [PMID: 30902563]
[9]
Guo F, Cui J. Anti-angiogenesis: Opening a new window for immunotherapy. Life Sciences 2020; 258: 118163.
[http://dx.doi.org/10.1016/j.lfs.2020.118163]
[10]
Khan KA, Kerbel RS. Improving immunotherapy outcomes with anti-angiogenic treatments and vice versa. Nat Rev Clin Oncol 2018; 15: 310-24.
[http://dx.doi.org/10.1038/nrclinonc.2018.9]
[11]
Albini A, Bruno A, Noonan DM, Mortara L. Contribution to tumor angiogenesis from innate immune cells within the tumor microenvironment: Implications for immunotherapy. Front Immunol 2018; 9: 527.
[http://dx.doi.org/10.3389/fimmu.2018.00527] [PMID: 29675018]
[12]
Viallard C, Larrivée B. Tumor angiogenesis and vascular normalization: Alternative therapeutic targets. Angiogenesis 2017; 20(4): 409-26.
[http://dx.doi.org/10.1007/s10456-017-9562-9] [PMID: 28660302]
[13]
Roy-Chowdhury S, Brown CK. Cytokines and tumor angiogenesis. In: Caligiuri MA, Lotze MT, Eds. Cytokines in the Genesis and Treatment of Cancer. Totowa, NJ: Humana Press 2007; pp. 245-66.
[http://dx.doi.org/10.1007/978-1-59745-455-1_14]
[14]
Montoya S, Soong D, Nguyen N, Affer M, Munamarty SP, Taylor J. Targeted therapies in cancer: To be or not to be, selective. Biomedicines 2021; 9(11): 1591.
[http://dx.doi.org/10.3390/biomedicines9111591] [PMID: 34829820]
[15]
Xia AL, Xu Y, Lu XJ. Cancer immunotherapy: Challenges and clinical applications. J Med Genet 2019; 56(1): 1-3.
[http://dx.doi.org/10.1136/jmedgenet-2018-105852] [PMID: 30464054]
[16]
Cassidy JW, Bruna A. Tumor Heterogeneity. In: Uthamanthil R, Tinkey P, Eds. Patient Derived Tumor Xenograft Models. Cambridge, USA: Academic Press 2017; pp. 37-55.
[http://dx.doi.org/10.1016/B978-0-12-804010-2.00004-7]
[17]
Gray M, Meehan J, Turnbull AK, et al. The importance of the tumor microenvironment and hypoxia in delivering a precision medicine approach to veterinary oncology. Front Vet Sci 2020; 7: 598338.
[http://dx.doi.org/10.3389/fvets.2020.598338] [PMID: 33282935]
[18]
Tang Z, Kang B, Li C, Chen T, Zhang Z. GEPIA2: An enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res 2019; 47(W1): W556-60.
[http://dx.doi.org/10.1093/nar/gkz430] [PMID: 31114875]
[19]
van der Maaten L, Hinton G. Viualizing data using t-SNE. J Mach Learn Res 2008; 9: 2579-605.
[20]
Zheng X, Li W, Ren L, et al. The sphingosine kinase-1/sphingosine-1-phosphate axis in cancer: Potential target for anticancer therapy. Pharmacol Ther 2019; 195: 85-99.
[http://dx.doi.org/10.1016/j.pharmthera.2018.10.011]
[21]
Sun X, Chen Q, Zhang L, Chen J, Zhang X. Exploration of prognostic biomarkers and therapeutic targets in the microenvironment of bladder cancer based on CXC chemokines. Math Biosci Eng 2021; 18(5): 6262-87.
[http://dx.doi.org/10.3934/mbe.2021313] [PMID: 34517533]
[22]
Wang Z, Yuan X, Jiao N, Zhu H, Zhang Y, Tong J. CDH13 and FLBN3 gene methylation are associated with poor prognosis in colorectal cancer. Pathol Oncol Res 2012; 18(2): 263-70.
[http://dx.doi.org/10.1007/s12253-011-9437-0]
[23]
Hatano K, Saigo C, Kito Y, Shibata T, Takeuchi T. Overexpression of JAG2 is related to poor outcomes in oral squamous cell carcinoma. Clin Exp Dent Res 2020; 6(2): 174-80.
[http://dx.doi.org/10.1002/cre2.267] [PMID: 32250571]
[24]
Yang S, Wang H, Qin C, Sun H, Han Y. Up-regulation of CXCL8 expression is associated with a poor prognosis and enhances tumor cell malignant behaviors in liver cancer. Biosci Rep 2020; 40(8): BSR20201169.
[http://dx.doi.org/10.1042/BSR20201169]
[25]
Wang H, Yin J, Hong Y, et al. SCG2 is a prognostic biomarker associated with immune infiltration and macrophage polarization in colorectal cancer. Front Cell Dev Biol 2022; 9: 795133.
[http://dx.doi.org/10.3389/fcell.2021.795133] [PMID: 35047505]
[26]
Bartoschek M, Pietras K. PDGF family function and prognostic value in tumor biology. Biochemical and Biophysical Research Communications 2018; 503(2): 984-90.
[http://dx.doi.org/10.1016/j.bbrc.2018.06.106]
[27]
Yuan L, Shu B, Chen L, et al. Overexpression of COL3A1 confers a poor prognosis in human bladder cancer identified by co-expression analysis. Oncotarget 2017; 8(41): 70508-20.
[http://dx.doi.org/10.18632/oncotarget.19733] [PMID: 29050298]
[28]
Connor EV, Saygin C, Braley C, et al. Thy-1 predicts poor prognosis and is associated with self-renewal in ovarian cancer. J Ovarian Res 2019; 12(1): 112.
[http://dx.doi.org/10.1186/s13048-019-0590-5] [PMID: 31735168]
[29]
Cheng G, Fan X, Hao M, Wang J, Zhou X, Sun X. Higher levels of TIMP-1 expression are associated with a poor prognosis in triple-negative breast cancer. Mol Cancer 2016; 15(1): 30.
[http://dx.doi.org/10.1186/s12943-016-0515-5] [PMID: 27130446]
[30]
Ding YL, Sun SF, Zhao GL. COL5A2 as a potential clinical biomarker for gastric cancer and renal metastasis. Medicine 2021; 100(7): e24561.
[http://dx.doi.org/10.1097/MD.0000000000024561] [PMID: 33607786]
[31]
Cong M, Yu T, Zhu L, Zhang J. TNNI3 expression and prognostic value and correlation with tumor-infiltrating immune cells in LIHC: A bioinformatics analysis. Research Square 2022.
[32]
Ferrari N, Mohammed ZMA, Nixon C, et al. Expression of RUNX1 correlates with poor patient prognosis in triple negative breast cancer. PLoS One 2014; 9(6): e100759.
[http://dx.doi.org/10.1371/journal.pone.0100759] [PMID: 24967588]
[33]
Li W, Han F, Fu M, Wang Z. High expression of VCAN is an independent predictor of poor prognosis in gastric cancer. J Int Med Res 2020; 48(1)
[http://dx.doi.org/10.1177/0300060519891271] [PMID: 31939331]
[34]
Gil M, Kim KE. Interleukin-18 is a prognostic biomarker correlated with CD8+ T cell and natural killer cell infiltration in skin cutaneous melanoma. J Clin Med 2019; 8(11): 1993.
[http://dx.doi.org/10.3390/jcm8111993]
[35]
Sun X, et al. OLR1 is a prognostic factor and correlated with immune infiltration in breast cancer. Int Immunopharmacol 2021; 101(Pt B): 108275.
[36]
Tang H, Chen J, Han X, Feng Y, Wang F. Upregulation of SPP1 is a marker for poor lung cancer prognosis and contributes to cancer progression and cisplatin resistance. Front Cell Dev Biol 2021; 9: 646390.
[http://dx.doi.org/10.3389/fcell.2021.646390] [PMID: 33996808]
[37]
Ge W, Shentu D, Wang Y, et al. A novel angiogenesis-based molecular signature related to prognosis and tumor immune interactions of pancreatic cancer. Front Cell Dev Biol 2022; 10: 1001606.
[http://dx.doi.org/10.3389/fcell.2022.1001606]
[38]
Chang ACM, Doherty J, Huschtscha LI, et al. STC1 expression is associated with tumor growth and metastasis in breast cancer. Clin Exp Metastasis 2015; 32(1): 15-27.
[http://dx.doi.org/10.1007/s10585-014-9687-9] [PMID: 25391215]
[39]
Oh HJ, Bae JM, Wen XY, Cho NY, Kim JH, Kang GH. Overexpression of POSTN in tumor stroma is a poor prognostic indicator of colorectal cancer. J Pathol Transl Med 2017; 51(3): 306-13.
[http://dx.doi.org/10.4132/jptm.2017.01.19] [PMID: 28407462]
[40]
Heppt M V, et al. MSX1-induced neural crest-like reprogramming promotes melanoma progression. J Invest Dermatol 2018; 138(1): 141-9.
[http://dx.doi.org/10.1016/j.jid.2017.05.038]
[41]
Zhu H, Zhao H, Wang J, et al. Potential prognosis index for m6A-related mRNA in cholangiocarcinoma. BMC Cancer 2022; 22(1): 620.
[http://dx.doi.org/10.1186/s12885-022-09665-3] [PMID: 35672673]
[42]
Gray RE, Harris GT. Renal cell carcinoma: Diagnosis and management. Am Fam Physician 2019; 99(3): 179-84.
[PMID: 30702258]
[43]
Rivera LB, Bergers G. Intertwined regulation of angiogenesis and immunity by myeloid cells. Trends Immunol 2015; 36(4): 240-9.
[http://dx.doi.org/10.1016/j.it.2015.02.005] [PMID: 25770923]
[44]
Trenti A, Tedesco S, Boscaro C, Trevisi L, Bolego C, Cignarella A. Estrogen, angiogenesis, immunity and cell metabolism: Solving the puzzle. Int J Mol Sci 2018; 19(3): 859.
[http://dx.doi.org/10.3390/ijms19030859] [PMID: 29543707]
[45]
Teicher B A. Antiangiogenic agents and targets: A perspective. Biochemical Pharmacology 2011; 81(1): 6-12.
[http://dx.doi.org/10.1016/j.bcp.2010.09.023]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy