Skip to main content
Top
Published in: Cancer Cell International 1/2019

Open Access 01-12-2019 | Colorectal Cancer | Primary research

A signature of hypoxia-related factors reveals functional dysregulation and robustly predicts clinical outcomes in stage I/II colorectal cancer patients

Authors: Yi-feng Zou, Yu-ming Rong, Ying-xin Tan, Jian Xiao, Zhao-liang Yu, Yu-feng Chen, Jia Ke, Cheng-hang Li, Xi Chen, Xiao-jian Wu, Ping Lan, Xu-tao Lin, Feng Gao

Published in: Cancer Cell International | Issue 1/2019

Login to get access

Abstract

Background

The hypoxic tumor microenvironment accelerates the invasion and migration of colorectal cancer (CRC) cells. The aim of this study was to develop and validate a hypoxia gene signature for predicting the outcome in stage I/II CRC patients that have limited therapeutic options.

Methods

The hypoxic gene signature (HGS) was constructed using transcriptomic data of 309 CRC patients with complete clinical information from the CIT microarray dataset. A total of 1877 CRC patients with complete prognostic information in six independent datasets were divided into a training cohort and two validation cohorts. Univariate and multivariate analyses were conducted to evaluate the prognostic value of HGS.

Results

The HGS consisted of 14 genes, and demarcated the CRC patients into the high- and low-risk groups. In all three cohorts, patients in the high-risk group had significantly worse disease free survival (DFS) compared with those in the low risk group (training cohort—HR = 4.35, 95% CI 2.30–8.23, P < 0.001; TCGA cohort—HR = 2.14, 95% CI 1.09–4.21, P = 0.024; meta-validation cohort—HR = 1.91, 95% CI 1.08–3.39, P = 0.024). Compared to Oncotype DX, HGS showed superior predictive outcome in the training cohort (C-index, 0.80 vs 0.65) and the validation cohort (C-index, 0.70 vs 0.61). Pathway analysis of the high- and low-HGS groups showed significant differences in the expression of genes involved in mTROC1, G2-M, mitosis, oxidative phosphorylation, MYC and PI3K–AKT–mTOR pathways (P < 0.005).

Conclusion

Hypoxic gene signature is a satisfactory prognostic model for early stage CRC patients, and the exact biological mechanism needs to be validated further.
Literature
1.
go back to reference Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.PubMedCrossRef Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68(6):394–424.PubMedCrossRef
2.
go back to reference van der Stok EP, Spaander MCW, Grunhagen DJ, Verhoef C, Kuipers EJ. Surveillance after curative treatment for colorectal cancer. Nat Rev Clin Oncol. 2017;14(5):297–315.PubMedCrossRef van der Stok EP, Spaander MCW, Grunhagen DJ, Verhoef C, Kuipers EJ. Surveillance after curative treatment for colorectal cancer. Nat Rev Clin Oncol. 2017;14(5):297–315.PubMedCrossRef
3.
go back to reference Brown JM, Giaccia AJ. The unique physiology of solid tumors: opportunities (and problems) for cancer therapy. Cancer Res. 1998;58(7):1408–16.PubMed Brown JM, Giaccia AJ. The unique physiology of solid tumors: opportunities (and problems) for cancer therapy. Cancer Res. 1998;58(7):1408–16.PubMed
4.
go back to reference Vaupel P, Mayer A, Hockel M. Tumor hypoxia and malignant progression. Methods Enzymol. 2004;381:335–54.PubMedCrossRef Vaupel P, Mayer A, Hockel M. Tumor hypoxia and malignant progression. Methods Enzymol. 2004;381:335–54.PubMedCrossRef
5.
go back to reference Yang L, Zhang W, Wang Y, et al. Hypoxia-induced miR-214 expression promotes tumour cell proliferation and migration by enhancing the Warburg effect in gastric carcinoma cells. Cancer Lett. 2018;414:44–56.PubMedCrossRef Yang L, Zhang W, Wang Y, et al. Hypoxia-induced miR-214 expression promotes tumour cell proliferation and migration by enhancing the Warburg effect in gastric carcinoma cells. Cancer Lett. 2018;414:44–56.PubMedCrossRef
6.
go back to reference Li H, Rokavec M, Jiang L, Horst D, Hermeking H. Antagonistic effects of p53 and HIF1A on microRNA-34a regulation of PPP1R11 and STAT3 and hypoxia-induced epithelial to mesenchymal transition in colorectal cancer cells. Gastroenterology. 2017;153(2):505–20.PubMedCrossRef Li H, Rokavec M, Jiang L, Horst D, Hermeking H. Antagonistic effects of p53 and HIF1A on microRNA-34a regulation of PPP1R11 and STAT3 and hypoxia-induced epithelial to mesenchymal transition in colorectal cancer cells. Gastroenterology. 2017;153(2):505–20.PubMedCrossRef
7.
go back to reference Kim CW, Oh ET, Kim JM, et al. Hypoxia-induced microRNA-590-5p promotes colorectal cancer progression by modulating matrix metalloproteinase activity. Cancer Lett. 2018;416:31–41.PubMedCrossRef Kim CW, Oh ET, Kim JM, et al. Hypoxia-induced microRNA-590-5p promotes colorectal cancer progression by modulating matrix metalloproteinase activity. Cancer Lett. 2018;416:31–41.PubMedCrossRef
8.
go back to reference Walsh JC, Lebedev A, Aten E, Madsen K, Marciano L, Kolb HC. The clinical importance of assessing tumor hypoxia: relationship of tumor hypoxia to prognosis and therapeutic opportunities. Antioxid Redox Signal. 2014;21(10):1516–54.PubMedPubMedCentralCrossRef Walsh JC, Lebedev A, Aten E, Madsen K, Marciano L, Kolb HC. The clinical importance of assessing tumor hypoxia: relationship of tumor hypoxia to prognosis and therapeutic opportunities. Antioxid Redox Signal. 2014;21(10):1516–54.PubMedPubMedCentralCrossRef
9.
go back to reference Li F, Mei H, Gao Y, et al. Co-delivery of oxygen and erlotinib by aptamer-modified liposomal complexes to reverse hypoxia-induced drug resistance in lung cancer. Biomaterials. 2017;145:56–71.PubMedCrossRef Li F, Mei H, Gao Y, et al. Co-delivery of oxygen and erlotinib by aptamer-modified liposomal complexes to reverse hypoxia-induced drug resistance in lung cancer. Biomaterials. 2017;145:56–71.PubMedCrossRef
10.
go back to reference Rechavi G, Brok-Simoni F, Ben-Bassat I, Ramot B. Spurious haemoglobinopathy. Lancet. 1986;1(8488):1035.PubMedCrossRef Rechavi G, Brok-Simoni F, Ben-Bassat I, Ramot B. Spurious haemoglobinopathy. Lancet. 1986;1(8488):1035.PubMedCrossRef
11.
go back to reference Betensky RA, Louis DN, Cairncross JG. Influence of unrecognized molecular heterogeneity on randomized clinical trials. J Clin Oncol. 2002;20(10):2495–9.PubMedCrossRef Betensky RA, Louis DN, Cairncross JG. Influence of unrecognized molecular heterogeneity on randomized clinical trials. J Clin Oncol. 2002;20(10):2495–9.PubMedCrossRef
12.
13.
go back to reference 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. 2005;102(43):15545–50.PubMedCrossRefPubMedCentral 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. 2005;102(43):15545–50.PubMedCrossRefPubMedCentral
14.
go back to reference Liberzon A, Subramanian A, Pinchback R, Thorvaldsdottir H, Tamayo P, Mesirov JP. Molecular signatures database (MSigDB) 3.0. Bioinformatics. 2011;27(12):1739–40.PubMedPubMedCentralCrossRef Liberzon A, Subramanian A, Pinchback R, Thorvaldsdottir H, Tamayo P, Mesirov JP. Molecular signatures database (MSigDB) 3.0. Bioinformatics. 2011;27(12):1739–40.PubMedPubMedCentralCrossRef
15.
go back to reference Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The molecular signatures database (MSigDB) hallmark gene set collection. Cell Syst. 2015;1(6):417–25.PubMedPubMedCentralCrossRef Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The molecular signatures database (MSigDB) hallmark gene set collection. Cell Syst. 2015;1(6):417–25.PubMedPubMedCentralCrossRef
16.
go back to reference Wang X, Terfve C, Rose JC, Markowetz F. HTSanalyzeR: an R/Bioconductor package for integrated network analysis of high-throughput screens. Bioinformatics. 2011;27(6):879–80.PubMedPubMedCentralCrossRef Wang X, Terfve C, Rose JC, Markowetz F. HTSanalyzeR: an R/Bioconductor package for integrated network analysis of high-throughput screens. Bioinformatics. 2011;27(6):879–80.PubMedPubMedCentralCrossRef
17.
go back to reference Markle B, May EJ, Majumdar AP. Do nutraceutics play a role in the prevention and treatment of colorectal cancer? Cancer Metastasis Rev. 2010;29(3):395–404.PubMedCrossRef Markle B, May EJ, Majumdar AP. Do nutraceutics play a role in the prevention and treatment of colorectal cancer? Cancer Metastasis Rev. 2010;29(3):395–404.PubMedCrossRef
18.
go back to reference Al-Temaimi RA, Tan TZ, Marafie MJ, Thiery JP, Quirke P, Al-Mulla F. Identification of 42 genes linked to stage II colorectal cancer metastatic relapse. Int J Mol Sci. 2016;17(5):598.PubMedCentralCrossRef Al-Temaimi RA, Tan TZ, Marafie MJ, Thiery JP, Quirke P, Al-Mulla F. Identification of 42 genes linked to stage II colorectal cancer metastatic relapse. Int J Mol Sci. 2016;17(5):598.PubMedCentralCrossRef
19.
go back to reference De Rosa M, Pace U, Rega D, et al. Genetics, diagnosis and management of colorectal cancer. Oncol Rep. 2015;34(3):1087–96.PubMedCrossRef De Rosa M, Pace U, Rega D, et al. Genetics, diagnosis and management of colorectal cancer. Oncol Rep. 2015;34(3):1087–96.PubMedCrossRef
20.
go back to reference Moniz S, Biddlestone J, Rocha S. Grow(2): the HIF system, energy homeostasis and the cell cycle. Histol Histopathol. 2014;29(5):589–600.PubMed Moniz S, Biddlestone J, Rocha S. Grow(2): the HIF system, energy homeostasis and the cell cycle. Histol Histopathol. 2014;29(5):589–600.PubMed
22.
go back to reference Vaupel P. The role of hypoxia-induced factors in tumor progression. Oncologist. 2004;9(Suppl 5):10–7.PubMedCrossRef Vaupel P. The role of hypoxia-induced factors in tumor progression. Oncologist. 2004;9(Suppl 5):10–7.PubMedCrossRef
23.
go back to reference Pahlman S, Mohlin S. Hypoxia and hypoxia-inducible factors in neuroblastoma. Cell Tissue Res. 2018;372(2):269–75.PubMedCrossRef Pahlman S, Mohlin S. Hypoxia and hypoxia-inducible factors in neuroblastoma. Cell Tissue Res. 2018;372(2):269–75.PubMedCrossRef
24.
go back to reference Rapisarda A, Uranchimeg B, Scudiero DA, et al. Identification of small molecule inhibitors of hypoxia-inducible factor 1 transcriptional activation pathway. Cancer Res. 2002;62(15):4316–24.PubMed Rapisarda A, Uranchimeg B, Scudiero DA, et al. Identification of small molecule inhibitors of hypoxia-inducible factor 1 transcriptional activation pathway. Cancer Res. 2002;62(15):4316–24.PubMed
25.
go back to reference Hockel M, Schlenger K, Aral B, Mitze M, Schaffer U, Vaupel P. Association between tumor hypoxia and malignant progression in advanced cancer of the uterine cervix. Cancer Res. 1996;56(19):4509–15.PubMed Hockel M, Schlenger K, Aral B, Mitze M, Schaffer U, Vaupel P. Association between tumor hypoxia and malignant progression in advanced cancer of the uterine cervix. Cancer Res. 1996;56(19):4509–15.PubMed
26.
go back to reference Sullivan R, Graham CH. Hypoxia-driven selection of the metastatic phenotype. Cancer Metastasis Rev. 2007;26(2):319–31.PubMedCrossRef Sullivan R, Graham CH. Hypoxia-driven selection of the metastatic phenotype. Cancer Metastasis Rev. 2007;26(2):319–31.PubMedCrossRef
28.
go back to reference Overgaard J. Clinical evaluation of nitroimidazoles as modifiers of hypoxia in solid tumors. Oncol Res. 1994;6(10–11):509–18.PubMed Overgaard J. Clinical evaluation of nitroimidazoles as modifiers of hypoxia in solid tumors. Oncol Res. 1994;6(10–11):509–18.PubMed
29.
go back to reference Overgaard J. Hypoxic modification of radiotherapy in squamous cell carcinoma of the head and neck—a systematic review and meta-analysis. Radiother Oncol. 2011;100(1):22–32.PubMedCrossRef Overgaard J. Hypoxic modification of radiotherapy in squamous cell carcinoma of the head and neck—a systematic review and meta-analysis. Radiother Oncol. 2011;100(1):22–32.PubMedCrossRef
31.
go back to reference Madhok BM, Yeluri S, Perry SL, Hughes TA, Jayne DG. Dichloroacetate induces apoptosis and cell-cycle arrest in colorectal cancer cells. Br J Cancer. 2010;102(12):1746–52.PubMedPubMedCentralCrossRef Madhok BM, Yeluri S, Perry SL, Hughes TA, Jayne DG. Dichloroacetate induces apoptosis and cell-cycle arrest in colorectal cancer cells. Br J Cancer. 2010;102(12):1746–52.PubMedPubMedCentralCrossRef
32.
go back to reference Oliveira A, Beyer G, Chugh R, et al. Triptolide abrogates growth of colon cancer and induces cell cycle arrest by inhibiting transcriptional activation of E2F. Lab Invest. 2015;95(6):648–59.PubMedPubMedCentralCrossRef Oliveira A, Beyer G, Chugh R, et al. Triptolide abrogates growth of colon cancer and induces cell cycle arrest by inhibiting transcriptional activation of E2F. Lab Invest. 2015;95(6):648–59.PubMedPubMedCentralCrossRef
33.
go back to reference Satoh K, Yachida S, Sugimoto M, et al. Global metabolic reprogramming of colorectal cancer occurs at adenoma stage and is induced by MYC. Proc Natl Acad Sci USA. 2017;114(37):E7697–706.PubMedCrossRefPubMedCentral Satoh K, Yachida S, Sugimoto M, et al. Global metabolic reprogramming of colorectal cancer occurs at adenoma stage and is induced by MYC. Proc Natl Acad Sci USA. 2017;114(37):E7697–706.PubMedCrossRefPubMedCentral
34.
go back to reference Sisinni L, Maddalena F, Condelli V, et al. TRAP1 controls cell cycle G2-M transition through the regulation of CDK1 and MAD2 expression/ubiquitination. J Pathol. 2017;243(1):123–34.PubMedCrossRef Sisinni L, Maddalena F, Condelli V, et al. TRAP1 controls cell cycle G2-M transition through the regulation of CDK1 and MAD2 expression/ubiquitination. J Pathol. 2017;243(1):123–34.PubMedCrossRef
35.
go back to reference Wang Y, Kuang H, Xue J, Liao L, Yin F, Zhou X. LncRNA AB073614 regulates proliferation and metastasis of colorectal cancer cells via the PI3K/AKT signaling pathway. Biomed Pharmacother. 2017;93:1230–7.PubMedCrossRef Wang Y, Kuang H, Xue J, Liao L, Yin F, Zhou X. LncRNA AB073614 regulates proliferation and metastasis of colorectal cancer cells via the PI3K/AKT signaling pathway. Biomed Pharmacother. 2017;93:1230–7.PubMedCrossRef
Metadata
Title
A signature of hypoxia-related factors reveals functional dysregulation and robustly predicts clinical outcomes in stage I/II colorectal cancer patients
Authors
Yi-feng Zou
Yu-ming Rong
Ying-xin Tan
Jian Xiao
Zhao-liang Yu
Yu-feng Chen
Jia Ke
Cheng-hang Li
Xi Chen
Xiao-jian Wu
Ping Lan
Xu-tao Lin
Feng Gao
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Cancer Cell International / Issue 1/2019
Electronic ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-019-0964-1

Other articles of this Issue 1/2019

Cancer Cell International 1/2019 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