Skip to main content
Top
Published in: Cancer Cell International 1/2021

Open Access 01-12-2021 | Primary research

Molecular mechanism study of HGF/c-MET pathway activation and immune regulation for a tumor diagnosis model

Authors: Zhibo Shen, Wenhua Xue, Yuanyuan Zheng, Qishun Geng, Le Wang, Zhirui Fan, Wenbin Wang, Ying Yue, Yunkai Zhai, Lifeng Li, Jie Zhao

Published in: Cancer Cell International | Issue 1/2021

Login to get access

Abstract

Background

Hepatocyte growth factor (HGF) binds to the c-mesenchymal-epithelial transition (C-MET) receptor and activates downstream signaling pathways, playing an essential role in the development of various cancers. Given the role of this signaling pathway, the primary therapeutic direction focuses on identifying and designing HGF inhibitors, antagonists and other molecules to block the binding of HGF to C-MET, thereby limiting the abnormal state of other downstream genes.

Methods

This study focuses on the analysis of immune-related genes and corresponding immune functions that are significantly associated with the HGF/c-MET pathway using transcriptome data from 11 solid tumors.

Results

We systematically analyzed 11 different cancers, including expression correlation, immune infiltration, tumor diagnosis and survival prognosis from HGF/c-MET pathway and immune regulation, two biological mechanisms having received extensive attention in cancer analysis.

Conclusion

We found that the HGF/c-MET pathway affected the tumor microenvironment mainly by interfering with expression levels of other genes. Immune infiltration is another critical factor involved in changes to the tumor microenvironment. The downstream immune-related genes activated by the HGF/c-MET pathway regulate immune-related pathways, which in turn affect the degree of infiltration of immune cells. Immune infiltration is significantly associated with cancer development and prognosis.
Appendix
Available only for authorised users
Literature
1.
go back to reference Konstorum A, Lowengrub JS. Activation of the HGF/c-Met axis in the tumor microenvironment: a multispecies model. J Theor Biol. 2018;439:86–99.CrossRef Konstorum A, Lowengrub JS. Activation of the HGF/c-Met axis in the tumor microenvironment: a multispecies model. J Theor Biol. 2018;439:86–99.CrossRef
2.
go back to reference Boromand N, Hasanzadeh M, ShahidSales S, Farazestanian M, Gharib M, Fiuji H, Behboodi N, Ghobadi N, Hassanian SM, Ferns GA, et al. Clinical and prognostic value of the C-Met/HGF signaling pathway in cervical cancer. J Cell Physiol. 2018;233(6):4490–6.CrossRef Boromand N, Hasanzadeh M, ShahidSales S, Farazestanian M, Gharib M, Fiuji H, Behboodi N, Ghobadi N, Hassanian SM, Ferns GA, et al. Clinical and prognostic value of the C-Met/HGF signaling pathway in cervical cancer. J Cell Physiol. 2018;233(6):4490–6.CrossRef
3.
go back to reference Granito A, Guidetti E, Gramantieri L. c-MET receptor tyrosine kinase as a molecular target in advanced hepatocellular carcinoma. J Hepatocell Carcinoma. 2015;2:29–38.PubMedPubMedCentral Granito A, Guidetti E, Gramantieri L. c-MET receptor tyrosine kinase as a molecular target in advanced hepatocellular carcinoma. J Hepatocell Carcinoma. 2015;2:29–38.PubMedPubMedCentral
5.
go back to reference Stanley A, Ashrafi GH, Seddon AM, Modjtahedi H. Synergistic effects of various Her inhibitors in combination with IGF-1R, C-MET and Src targeting agents in breast cancer cell lines. Sci Rep. 2017;7(1):3964.CrossRef Stanley A, Ashrafi GH, Seddon AM, Modjtahedi H. Synergistic effects of various Her inhibitors in combination with IGF-1R, C-MET and Src targeting agents in breast cancer cell lines. Sci Rep. 2017;7(1):3964.CrossRef
6.
go back to reference Lam BQ, Dai L, Qin Z. The role of HGF/c-MET signaling pathway in lymphoma. J Hematol Oncol. 2016;9(1):135.CrossRef Lam BQ, Dai L, Qin Z. The role of HGF/c-MET signaling pathway in lymphoma. J Hematol Oncol. 2016;9(1):135.CrossRef
7.
go back to reference Krause DS, Van Etten RA. Tyrosine kinases as targets for cancer therapy. N Engl J Med. 2005;353(2):172–87.CrossRef Krause DS, Van Etten RA. Tyrosine kinases as targets for cancer therapy. N Engl J Med. 2005;353(2):172–87.CrossRef
9.
go back to reference Hu CT, Wu JR, Cheng CC, Wu WS. The therapeutic targeting of HGF/c-Met signaling in hepatocellular carcinoma: alternative approaches. Cancers. 2017;9(6):58.CrossRef Hu CT, Wu JR, Cheng CC, Wu WS. The therapeutic targeting of HGF/c-Met signaling in hepatocellular carcinoma: alternative approaches. Cancers. 2017;9(6):58.CrossRef
10.
go back to reference Bradley CA, Salto-Tellez M, Laurent-Puig P, Bardelli A, Rolfo C, Tabernero J, Khawaja HA, Lawler M, Johnston PG, Van Schaeybroeck S, et al. Targeting c-MET in gastrointestinal tumours: rationale, opportunities and challenges. Nat Rev Clin Oncol. 2017;14(9):562–76.CrossRef Bradley CA, Salto-Tellez M, Laurent-Puig P, Bardelli A, Rolfo C, Tabernero J, Khawaja HA, Lawler M, Johnston PG, Van Schaeybroeck S, et al. Targeting c-MET in gastrointestinal tumours: rationale, opportunities and challenges. Nat Rev Clin Oncol. 2017;14(9):562–76.CrossRef
11.
go back to reference Xu X, Zhu Y, Liang Z, Li S, Xu X, Wang X, Wu J, Hu Z, Meng S, Liu B, et al. c-Met and CREB1 are involved in miR-433-mediated inhibition of the epithelial-mesenchymal transition in bladder cancer by regulating Akt/GSK-3beta/Snail signaling. Cell Death Dis. 2016;7:e2088.CrossRef Xu X, Zhu Y, Liang Z, Li S, Xu X, Wang X, Wu J, Hu Z, Meng S, Liu B, et al. c-Met and CREB1 are involved in miR-433-mediated inhibition of the epithelial-mesenchymal transition in bladder cancer by regulating Akt/GSK-3beta/Snail signaling. Cell Death Dis. 2016;7:e2088.CrossRef
12.
go back to reference Furge KA, Zhang YW, Vande Woude GF. Met receptor tyrosine kinase: enhanced signaling through adapter proteins. Oncogene. 2000;19(49):5582–9.CrossRef Furge KA, Zhang YW, Vande Woude GF. Met receptor tyrosine kinase: enhanced signaling through adapter proteins. Oncogene. 2000;19(49):5582–9.CrossRef
13.
go back to reference Wang W, Dong J, Wang M, Yao S, Tian X, Cui X, Fu S, Zhang S. miR-148a-3p suppresses epithelial ovarian cancer progression primarily by targeting c-Met. Oncol Lett. 2018;15(5):6131–6.PubMedPubMedCentral Wang W, Dong J, Wang M, Yao S, Tian X, Cui X, Fu S, Zhang S. miR-148a-3p suppresses epithelial ovarian cancer progression primarily by targeting c-Met. Oncol Lett. 2018;15(5):6131–6.PubMedPubMedCentral
14.
go back to reference Demkova L, Kucerova L. Role of the HGF/c-MET tyrosine kinase inhibitors in metastasic melanoma. Mol cancer. 2018;17(1):26.CrossRef Demkova L, Kucerova L. Role of the HGF/c-MET tyrosine kinase inhibitors in metastasic melanoma. Mol cancer. 2018;17(1):26.CrossRef
15.
go back to reference Pasquini G, Giaccone G. C-MET inhibitors for advanced non-small cell lung cancer. Expert Opin Investig Drugs. 2018;27(4):363–75.CrossRef Pasquini G, Giaccone G. C-MET inhibitors for advanced non-small cell lung cancer. Expert Opin Investig Drugs. 2018;27(4):363–75.CrossRef
16.
go back to reference Christensen JG, Burrows J, Salgia R. c-Met as a target for human cancer and characterization of inhibitors for therapeutic intervention. Cancer Lett. 2005;225(1):1–26.CrossRef Christensen JG, Burrows J, Salgia R. c-Met as a target for human cancer and characterization of inhibitors for therapeutic intervention. Cancer Lett. 2005;225(1):1–26.CrossRef
17.
go back to reference Goc J, Fridman WH, Sautes-Fridman C, Dieu-Nosjean MC. Characteristics of tertiary lymphoid structures in primary cancers. Oncoimmunology. 2013;2(12):e26836.CrossRef Goc J, Fridman WH, Sautes-Fridman C, Dieu-Nosjean MC. Characteristics of tertiary lymphoid structures in primary cancers. Oncoimmunology. 2013;2(12):e26836.CrossRef
18.
go back to reference Pages F, Berger A, Camus M, Sanchez-Cabo F, Costes A, Molidor R, Mlecnik B, Kirilovsky A, Nilsson M, Damotte D, et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med. 2005;353(25):2654–66.CrossRef Pages F, Berger A, Camus M, Sanchez-Cabo F, Costes A, Molidor R, Mlecnik B, Kirilovsky A, Nilsson M, Damotte D, et al. Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med. 2005;353(25):2654–66.CrossRef
19.
go back to reference Naito Y, Saito K, Shiiba K, Ohuchi A, Saigenji K, Nagura H, Ohtani H. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res. 1998;58(16):3491–4.PubMed Naito Y, Saito K, Shiiba K, Ohuchi A, Saigenji K, Nagura H, Ohtani H. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res. 1998;58(16):3491–4.PubMed
20.
go back to reference Somekh J, Shen-Orr SS, Kohane IS. Batch correction evaluation framework using a-priori gene-gene associations: applied to the GTEx dataset. BMC bioinformatics. 2019;20(1):268.CrossRef Somekh J, Shen-Orr SS, Kohane IS. Batch correction evaluation framework using a-priori gene-gene associations: applied to the GTEx dataset. BMC bioinformatics. 2019;20(1):268.CrossRef
21.
go back to reference Bhattacharya S, Andorf S, Gomes L, Dunn P, Schaefer H, Pontius J, Berger P, Desborough V, Smith T, Campbell J, et al. ImmPort: disseminating data to the public for the future of immunology. Immunol Res. 2014;58(2–3):234–9.CrossRef Bhattacharya S, Andorf S, Gomes L, Dunn P, Schaefer H, Pontius J, Berger P, Desborough V, Smith T, Campbell J, et al. ImmPort: disseminating data to the public for the future of immunology. Immunol Res. 2014;58(2–3):234–9.CrossRef
22.
go back to reference Costa-Silva J, Domingues D, Lopes FM. RNA-Seq differential expression analysis: An extended review and a software tool. PloS One. 2017;12(12):e0190152.CrossRef Costa-Silva J, Domingues D, Lopes FM. RNA-Seq differential expression analysis: An extended review and a software tool. PloS One. 2017;12(12):e0190152.CrossRef
23.
go back to reference Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics. 2012;16(5):284–7.CrossRef Yu G, Wang LG, Han Y, He QY. clusterProfiler: an R package for comparing biological themes among gene clusters. Omics. 2012;16(5):284–7.CrossRef
24.
go back to reference Wu T, Wang Y, Jiang R, Lu X, Tian J. A pathways-based prediction model for classifying breast cancer subtypes. Oncotarget. 2017;8(35):58809–22.CrossRef Wu T, Wang Y, Jiang R, Lu X, Tian J. A pathways-based prediction model for classifying breast cancer subtypes. Oncotarget. 2017;8(35):58809–22.CrossRef
25.
go back to reference Chen B, Khodadoust MS, Liu CL, Newman AM, Alizadeh AA. Profiling Tumor Infiltrating Immune Cells with CIBERSORT. Methods Mol Biol. 2018;1711:243–59.CrossRef Chen B, Khodadoust MS, Liu CL, Newman AM, Alizadeh AA. Profiling Tumor Infiltrating Immune Cells with CIBERSORT. Methods Mol Biol. 2018;1711:243–59.CrossRef
26.
go back to reference Scalabrini Sampaio G, Vallim Filho ARA, da Santos Silva L, da Augusto Silva L. Prediction of motor failure time using an artificial neural network. Sensors. 2019;19(19):4342.CrossRef Scalabrini Sampaio G, Vallim Filho ARA, da Santos Silva L, da Augusto Silva L. Prediction of motor failure time using an artificial neural network. Sensors. 2019;19(19):4342.CrossRef
27.
go back to reference Williams C, Lewsey JD, Briggs AH, Mackay DF. Cost-effectiveness Analysis in R using a multi-state modeling survival analysis framework: a tutorial. Med Decis Making. 2017;37(4):340–52.CrossRef Williams C, Lewsey JD, Briggs AH, Mackay DF. Cost-effectiveness Analysis in R using a multi-state modeling survival analysis framework: a tutorial. Med Decis Making. 2017;37(4):340–52.CrossRef
28.
go back to reference Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331(6024):1565–70.CrossRef Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331(6024):1565–70.CrossRef
Metadata
Title
Molecular mechanism study of HGF/c-MET pathway activation and immune regulation for a tumor diagnosis model
Authors
Zhibo Shen
Wenhua Xue
Yuanyuan Zheng
Qishun Geng
Le Wang
Zhirui Fan
Wenbin Wang
Ying Yue
Yunkai Zhai
Lifeng Li
Jie Zhao
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2021
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-021-02051-2

Other articles of this Issue 1/2021

Cancer Cell International 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine