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

01-12-2021 | Cetuximab | Primary research

Identification of key genes involved in tumor immune cell infiltration and cetuximab resistance in colorectal cancer

Authors: Li Liang, Mengling Liu, Xun Sun, Yitao Yuan, Ke Peng, Khalid Rashid, Yiyi Yu, Yuehong Cui, Yanjie Chen, Tianshu Liu

Published in: Cancer Cell International | Issue 1/2021

Login to get access

Abstract

Background

The anti-epidermal growth factor receptor (EGFR) antibody introduces adaptable variations to the transcriptome and triggers tumor immune infiltration, resulting in colorectal cancer (CRC) treatment resistance. We intended to identify genes that play essential roles in cetuximab resistance and tumor immune cell infiltration.

Methods

A cetuximab-resistant CACO2 cellular model was established, and its transcriptome variations were detected by microarray. Meanwhile, public data from the Gene Expression Omnibus and The Cancer Genome Atlas (TCGA) database were downloaded. Integrated bioinformatics analysis was applied to detect differentially expressed genes (DEGs) between the cetuximab-resistant and the cetuximab-sensitive groups. Then, we investigated correlations between DEGs and immune cell infiltration. The DEGs from bioinformatics analysis were further validated in vitro and in clinical samples.

Results

We identified 732 upregulated and 1259 downregulated DEGs in the induced cellular model. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses, along with Gene Set Enrichment Analysis and Gene Set Variation Analysis, indicated the functions of the DEGs. Together with GSE59857 and GSE5841, 12 common DEGs (SATB-2, AKR1B10, ADH1A, ADH1C, MYB, ATP10B, CDX-2, FAR2, EPHB2, SLC26A3, ORP-1, VAV3) were identified and their predictive values of cetuximab treatment were validated in GSE56386. In online Genomics of Drug Sensitivity in Cancer (GDSC) database, nine of twelve DEGs were recognized in the protein-protein (PPI) network. Based on the transcriptome profiles of CRC samples in TCGA and using Tumor Immune Estimation Resource Version 2.0, we bioinformatically determined that SATB-2, ORP-1, MYB, and CDX-2 expressions were associated with intensive infiltration of B cell, CD4+ T cell, CD8+ T cell and macrophage, which was then validated the correlation in clinical samples by immunohistochemistry. We found that SATB-2, ORP-1, MYB, and CDX-2 were downregulated in vitro with cetuximab treatment. Clinically, patients with advanced CRC and high ORP-1 expression exhibited a longer progression-free survival time when they were treated with anti-EGFR therapy than those with low ORP-1 expression.

Conclusions

SATB-2, ORP-1, MYB, and CDX-2 were related to cetuximab sensitivity as well as enhanced tumor immune cell infiltration in patients with CRC.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115–32.CrossRefPubMed Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ, He J. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115–32.CrossRefPubMed
3.
go back to reference Heinemann V, von Weikersthal LF, Decker T, Kiani A, Vehling-Kaiser U, Al-Batran SE, Heintges T, Lerchenmuller C, Kahl C, Seipelt G, et al. FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for patients with metastatic colorectal cancer (FIRE-3): a randomised, open-label, phase 3 trial. Lancet Oncol. 2014;15(10):1065–75.PubMedCrossRef Heinemann V, von Weikersthal LF, Decker T, Kiani A, Vehling-Kaiser U, Al-Batran SE, Heintges T, Lerchenmuller C, Kahl C, Seipelt G, et al. FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for patients with metastatic colorectal cancer (FIRE-3): a randomised, open-label, phase 3 trial. Lancet Oncol. 2014;15(10):1065–75.PubMedCrossRef
4.
go back to reference Avallone A, Budillon A. Impact of subsequent therapies on outcome of the FIRE-3/AIO KRK0306 trial. J Clin Oncol. 2016;34(13):1564.PubMedCrossRef Avallone A, Budillon A. Impact of subsequent therapies on outcome of the FIRE-3/AIO KRK0306 trial. J Clin Oncol. 2016;34(13):1564.PubMedCrossRef
5.
go back to reference Primrose J, Falk S, Finch-Jones M, Valle J, O’Reilly D, Siriwardena A, Hornbuckle J, Peterson M, Rees M, Iveson T, et al. Systemic chemotherapy with or without cetuximab in patients with resectable colorectal liver metastasis: the New EPOC randomised controlled trial. Lancet Oncol. 2014;15(6):601–11.PubMedCrossRef Primrose J, Falk S, Finch-Jones M, Valle J, O’Reilly D, Siriwardena A, Hornbuckle J, Peterson M, Rees M, Iveson T, et al. Systemic chemotherapy with or without cetuximab in patients with resectable colorectal liver metastasis: the New EPOC randomised controlled trial. Lancet Oncol. 2014;15(6):601–11.PubMedCrossRef
6.
go back to reference Levi FA, Boige V, Hebbar M, Smith D, Lepere C, Focan C, Karaboue A, Guimbaud R, Carvalho C, Tumolo S, et al. Conversion to resection of liver metastases from colorectal cancer with hepatic artery infusion of combined chemotherapy and systemic cetuximab in multicenter trial OPTILIV. Ann Oncol. 2016;27(2):267–74.PubMedCrossRef Levi FA, Boige V, Hebbar M, Smith D, Lepere C, Focan C, Karaboue A, Guimbaud R, Carvalho C, Tumolo S, et al. Conversion to resection of liver metastases from colorectal cancer with hepatic artery infusion of combined chemotherapy and systemic cetuximab in multicenter trial OPTILIV. Ann Oncol. 2016;27(2):267–74.PubMedCrossRef
7.
go back to reference Bridgewater JA, Pugh SA, Maishman T, Eminton Z, Mellor J, Whitehead A, Stanton L, Radford M, Corkhill A, Griffiths GO, et al. Systemic chemotherapy with or without cetuximab in patients with resectable colorectal liver metastasis (New EPOC): long-term results of a multicentre, randomised, controlled, phase 3 trial. Lancet Oncol. 2020;21(3):398–411.PubMedPubMedCentralCrossRef Bridgewater JA, Pugh SA, Maishman T, Eminton Z, Mellor J, Whitehead A, Stanton L, Radford M, Corkhill A, Griffiths GO, et al. Systemic chemotherapy with or without cetuximab in patients with resectable colorectal liver metastasis (New EPOC): long-term results of a multicentre, randomised, controlled, phase 3 trial. Lancet Oncol. 2020;21(3):398–411.PubMedPubMedCentralCrossRef
8.
go back to reference Garattini S, Torri V, Floriani I. Cetuximab for metastatic colorectal cancer. N Engl J Med. 2009;361(1):96. author reply 96–97.PubMed Garattini S, Torri V, Floriani I. Cetuximab for metastatic colorectal cancer. N Engl J Med. 2009;361(1):96. author reply 96–97.PubMed
9.
go back to reference Venook AP, Niedzwiecki D, Lenz HJ, Innocenti F, Fruth B, Meyerhardt JA, Schrag D, Greene C, O’Neil BH, Atkins JN, et al. Effect of first-line chemotherapy combined with cetuximab or bevacizumab on overall survival in patients with KRAS wild-type advanced or metastatic colorectal cancer: a randomized clinical trial. JAMA. 2017;317(23):2392–401.PubMedPubMedCentralCrossRef Venook AP, Niedzwiecki D, Lenz HJ, Innocenti F, Fruth B, Meyerhardt JA, Schrag D, Greene C, O’Neil BH, Atkins JN, et al. Effect of first-line chemotherapy combined with cetuximab or bevacizumab on overall survival in patients with KRAS wild-type advanced or metastatic colorectal cancer: a randomized clinical trial. JAMA. 2017;317(23):2392–401.PubMedPubMedCentralCrossRef
10.
go back to reference Adam R, Aloia T, Levi F, Wicherts DA, de Haas RJ, Paule B, Bralet MP, Bouchahda M, Machover D, Ducreux M, et al. Hepatic resection after rescue cetuximab treatment for colorectal liver metastases previously refractory to conventional systemic therapy. J Clin Oncol. 2007;25(29):4593–602.PubMedCrossRef Adam R, Aloia T, Levi F, Wicherts DA, de Haas RJ, Paule B, Bralet MP, Bouchahda M, Machover D, Ducreux M, et al. Hepatic resection after rescue cetuximab treatment for colorectal liver metastases previously refractory to conventional systemic therapy. J Clin Oncol. 2007;25(29):4593–602.PubMedCrossRef
11.
go back to reference Bregni G, Sciallero S, Sobrero A. HER2 amplification and anti-EGFR sensitivity in advanced colorectal cancer. JAMA Oncol. 2019;5(5):605–6.PubMedCrossRef Bregni G, Sciallero S, Sobrero A. HER2 amplification and anti-EGFR sensitivity in advanced colorectal cancer. JAMA Oncol. 2019;5(5):605–6.PubMedCrossRef
12.
go back to reference Waddell T. Targeting EGFR in colorectal cancer: beyond KRAS exon 2. Lancet Oncol. 2014;15(6):540–1.PubMedCrossRef Waddell T. Targeting EGFR in colorectal cancer: beyond KRAS exon 2. Lancet Oncol. 2014;15(6):540–1.PubMedCrossRef
13.
go back to reference Diaz LA Jr, Williams RT, Wu J, Kinde I, Hecht JR, Berlin J, Allen B, Bozic I, Reiter JG, Nowak MA, et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature. 2012;486(7404):537–40.PubMedPubMedCentralCrossRef Diaz LA Jr, Williams RT, Wu J, Kinde I, Hecht JR, Berlin J, Allen B, Bozic I, Reiter JG, Nowak MA, et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature. 2012;486(7404):537–40.PubMedPubMedCentralCrossRef
14.
go back to reference Sahin IH, Oktay AA. EGFR signalling and oncogenic pathway signature in colorectal cancer. Gut. 2013;62(11):1670.PubMedCrossRef Sahin IH, Oktay AA. EGFR signalling and oncogenic pathway signature in colorectal cancer. Gut. 2013;62(11):1670.PubMedCrossRef
15.
go back to reference Rachiglio AM, Lambiase M, Fenizia F, Roma C, Cardone C, Iannaccone A, De Luca A, Carotenuto M, Frezzetti D, Martinelli E, et al. Genomic profiling of KRAS/NRAS/BRAF/PIK3CA wild-type metastatic colorectal cancer patients reveals novel mutations in genes potentially associated with resistance to anti-EGFR agents. Cancers (Basel). 2019;11(6):859.CrossRef Rachiglio AM, Lambiase M, Fenizia F, Roma C, Cardone C, Iannaccone A, De Luca A, Carotenuto M, Frezzetti D, Martinelli E, et al. Genomic profiling of KRAS/NRAS/BRAF/PIK3CA wild-type metastatic colorectal cancer patients reveals novel mutations in genes potentially associated with resistance to anti-EGFR agents. Cancers (Basel). 2019;11(6):859.CrossRef
16.
go back to reference Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 2013;369(11):1023–34.PubMedCrossRef Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, Humblet Y, Bodoky G, Cunningham D, Jassem J, et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 2013;369(11):1023–34.PubMedCrossRef
17.
go back to reference Sorich MJ, Wiese MD, Rowland A, Kichenadasse G, McKinnon RA, Karapetis CS. Extended RAS mutations and anti-EGFR monoclonal antibody survival benefit in metastatic colorectal cancer: a meta-analysis of randomized, controlled trials. Ann Oncol. 2015;26(1):13–21.PubMedCrossRef Sorich MJ, Wiese MD, Rowland A, Kichenadasse G, McKinnon RA, Karapetis CS. Extended RAS mutations and anti-EGFR monoclonal antibody survival benefit in metastatic colorectal cancer: a meta-analysis of randomized, controlled trials. Ann Oncol. 2015;26(1):13–21.PubMedCrossRef
18.
go back to reference Bertotti A, Papp E, Jones S, Adleff V, Anagnostou V, Lupo B, Sausen M, Phallen J, Hruban CA, Tokheim C, et al. The genomic landscape of response to EGFR blockade in colorectal cancer. Nature. 2015;526(7572):263–7.PubMedPubMedCentralCrossRef Bertotti A, Papp E, Jones S, Adleff V, Anagnostou V, Lupo B, Sausen M, Phallen J, Hruban CA, Tokheim C, et al. The genomic landscape of response to EGFR blockade in colorectal cancer. Nature. 2015;526(7572):263–7.PubMedPubMedCentralCrossRef
19.
go back to reference Misale S, Di Nicolantonio F, Sartore-Bianchi A, Siena S, Bardelli A. Resistance to anti-EGFR therapy in colorectal cancer: from heterogeneity to convergent evolution. Cancer Discov. 2014;4(11):1269–80.PubMedCrossRef Misale S, Di Nicolantonio F, Sartore-Bianchi A, Siena S, Bardelli A. Resistance to anti-EGFR therapy in colorectal cancer: from heterogeneity to convergent evolution. Cancer Discov. 2014;4(11):1269–80.PubMedCrossRef
20.
go back to reference Woolston A, Khan K, Cunningham D, Gerlinger M. Genomic and transcriptomic determinants of therapy resistance and immune landscape evolution during anti-EGFR treatment in colorectal cancer. Cancer Cell. 2019;36(1):35-50 e39.PubMedPubMedCentralCrossRef Woolston A, Khan K, Cunningham D, Gerlinger M. Genomic and transcriptomic determinants of therapy resistance and immune landscape evolution during anti-EGFR treatment in colorectal cancer. Cancer Cell. 2019;36(1):35-50 e39.PubMedPubMedCentralCrossRef
21.
go back to reference Pozzi C, Cuomo A, Spadoni I, Magni E, Silvola A, Conte A, Sigismund S, Ravenda PS, Bonaldi T, Zampino MG, et al. The EGFR-specific antibody cetuximab combined with chemotherapy triggers immunogenic cell death. Nat Med. 2016;22(6):624–31.PubMedCrossRef Pozzi C, Cuomo A, Spadoni I, Magni E, Silvola A, Conte A, Sigismund S, Ravenda PS, Bonaldi T, Zampino MG, et al. The EGFR-specific antibody cetuximab combined with chemotherapy triggers immunogenic cell death. Nat Med. 2016;22(6):624–31.PubMedCrossRef
22.
go back to reference Giordano GR, Porras A, Pancione AM. Immune resistance and EGFR antagonists in colorectal cancer. Cancers (Basel). 2019;11(8):1089.CrossRef Giordano GR, Porras A, Pancione AM. Immune resistance and EGFR antagonists in colorectal cancer. Cancers (Basel). 2019;11(8):1089.CrossRef
23.
go back to reference Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47.PubMedPubMedCentralCrossRef Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47.PubMedPubMedCentralCrossRef
24.
go back to reference Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, Benner C, Chanda SK. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10(1):1523.PubMedPubMedCentralCrossRef Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, Benner C, Chanda SK. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10(1):1523.PubMedPubMedCentralCrossRef
25.
go back to reference Subramanian A, Kuehn H, Gould J, Tamayo P, Mesirov JP. GSEA-P: a desktop application for Gene Set Enrichment Analysis. Bioinformatics. 2007;23(23):3251–3.PubMedCrossRef Subramanian A, Kuehn H, Gould J, Tamayo P, Mesirov JP. GSEA-P: a desktop application for Gene Set Enrichment Analysis. Bioinformatics. 2007;23(23):3251–3.PubMedCrossRef
26.
go back to reference Ferreira MR, Santos GA, Biagi CA, Silva Junior WA, Zambuzzi WF. GSVA score reveals molecular signatures from transcriptomes for biomaterials comparison. J Biomed Mater Res A 2020. Ferreira MR, Santos GA, Biagi CA, Silva Junior WA, Zambuzzi WF. GSVA score reveals molecular signatures from transcriptomes for biomaterials comparison. J Biomed Mater Res A 2020.
27.
go back to reference Yang W, Soares J, Greninger P, Edelman EJ, Lightfoot H, Forbes S, Bindal N, Beare D, Smith JA, Thompson IR, et al. Genomics of Drug Sensitivity in Cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 2013;41:D955-61 Database issue.PubMedCrossRef Yang W, Soares J, Greninger P, Edelman EJ, Lightfoot H, Forbes S, Bindal N, Beare D, Smith JA, Thompson IR, et al. Genomics of Drug Sensitivity in Cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells. Nucleic Acids Res. 2013;41:D955-61 Database issue.PubMedCrossRef
28.
go back to reference Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, Pyysalo S, Doncheva NT, Legeay M, Fang T, Bork P, et al. The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021;49(D1):D605–12.PubMedCrossRef Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, Pyysalo S, Doncheva NT, Legeay M, Fang T, Bork P, et al. The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021;49(D1):D605–12.PubMedCrossRef
29.
30.
go back to reference Li T, Fu J, Zeng Z, Cohen D, Li J, Chen Q, Li B, Liu XS. TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res. 2020;48(W1):W509–14.PubMedPubMedCentralCrossRef Li T, Fu J, Zeng Z, Cohen D, Li J, Chen Q, Li B, Liu XS. TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res. 2020;48(W1):W509–14.PubMedPubMedCentralCrossRef
31.
go back to reference Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, Li B, Liu XS. TIMER: a web server for comprehensive analysis of tumor-infiltrating immune cells. Cancer Res. 2017;77(21):e108–10.PubMedPubMedCentralCrossRef Li T, Fan J, Wang B, Traugh N, Chen Q, Liu JS, Li B, Liu XS. TIMER: a web server for comprehensive analysis of tumor-infiltrating immune cells. Cancer Res. 2017;77(21):e108–10.PubMedPubMedCentralCrossRef
32.
go back to reference Garvey CM, Lau R, Sanchez A, Sun RX, Fong EJ, Doche ME, Chen O, Jusuf A, Lenz HJ, Larson B, et al. Anti-EGFR therapy induces EGF secretion by cancer-associated fibroblasts to confer colorectal cancer chemoresistance. Cancers (Basel). 2020;12(6):1393.CrossRef Garvey CM, Lau R, Sanchez A, Sun RX, Fong EJ, Doche ME, Chen O, Jusuf A, Lenz HJ, Larson B, et al. Anti-EGFR therapy induces EGF secretion by cancer-associated fibroblasts to confer colorectal cancer chemoresistance. Cancers (Basel). 2020;12(6):1393.CrossRef
33.
go back to reference Mansour MA, Hyodo T, Ito S, Kurita K, Kokuryo T, Uehara K, Nagino M, Takahashi M, Hamaguchi M, Senga T. SATB2 suppresses the progression of colorectal cancer cells via inactivation of MEK5/ERK5 signaling. FEBS J. 2015;282(8):1394–405.PubMedCrossRef Mansour MA, Hyodo T, Ito S, Kurita K, Kokuryo T, Uehara K, Nagino M, Takahashi M, Hamaguchi M, Senga T. SATB2 suppresses the progression of colorectal cancer cells via inactivation of MEK5/ERK5 signaling. FEBS J. 2015;282(8):1394–405.PubMedCrossRef
34.
go back to reference Xu M, Xu X, Pan B, Chen X, Lin K, Zeng K, Liu X, Xu T, Sun L, Qin J, et al. LncRNA SATB2-AS1 inhibits tumor metastasis and affects the tumor immune cell microenvironment in colorectal cancer by regulating SATB2. Mol Cancer. 2019;18(1):135.PubMedPubMedCentralCrossRef Xu M, Xu X, Pan B, Chen X, Lin K, Zeng K, Liu X, Xu T, Sun L, Qin J, et al. LncRNA SATB2-AS1 inhibits tumor metastasis and affects the tumor immune cell microenvironment in colorectal cancer by regulating SATB2. Mol Cancer. 2019;18(1):135.PubMedPubMedCentralCrossRef
35.
go back to reference Johansson M, Lehto M, Tanhuanpaa K, Cover TL, Olkkonen VM. The oxysterol-binding protein homologue ORP1L interacts with Rab7 and alters functional properties of late endocytic compartments. Mol Biol Cell. 2005;16(12):5480–92.PubMedPubMedCentralCrossRef Johansson M, Lehto M, Tanhuanpaa K, Cover TL, Olkkonen VM. The oxysterol-binding protein homologue ORP1L interacts with Rab7 and alters functional properties of late endocytic compartments. Mol Biol Cell. 2005;16(12):5480–92.PubMedPubMedCentralCrossRef
36.
go back to reference Suchanek M, Hynynen R, Wohlfahrt G, Lehto M, Johansson M, Saarinen H, Radzikowska A, Thiele C, Olkkonen VM. The mammalian oxysterol-binding protein-related proteins (ORPs) bind 25-hydroxycholesterol in an evolutionarily conserved pocket. Biochem J. 2007;405(3):473–80.PubMedPubMedCentralCrossRef Suchanek M, Hynynen R, Wohlfahrt G, Lehto M, Johansson M, Saarinen H, Radzikowska A, Thiele C, Olkkonen VM. The mammalian oxysterol-binding protein-related proteins (ORPs) bind 25-hydroxycholesterol in an evolutionarily conserved pocket. Biochem J. 2007;405(3):473–80.PubMedPubMedCentralCrossRef
37.
go back to reference Motazacker MM, Pirhonen J, van Capelleveen JC, Weber-Boyvat M, Kuivenhoven JA, Shah S, Hovingh GK, Metso J, Li S, Ikonen E, et al. A loss-of-function variant in OSBPL1A predisposes to low plasma HDL cholesterol levels and impaired cholesterol efflux capacity. Atherosclerosis. 2016;249:140–7.PubMedCrossRef Motazacker MM, Pirhonen J, van Capelleveen JC, Weber-Boyvat M, Kuivenhoven JA, Shah S, Hovingh GK, Metso J, Li S, Ikonen E, et al. A loss-of-function variant in OSBPL1A predisposes to low plasma HDL cholesterol levels and impaired cholesterol efflux capacity. Atherosclerosis. 2016;249:140–7.PubMedCrossRef
38.
go back to reference Tichy M, Knopfova L, Jarkovsky J, Pekarcikova L, Veverkova L, Vlcek P, Katolicka J, Capov I, Hermanova M, Smarda J, et al. Overexpression of c-Myb is associated with suppression of distant metastases in colorectal carcinoma. Tumour Biol. 2016;37(8):10723–9.PubMedCrossRef Tichy M, Knopfova L, Jarkovsky J, Pekarcikova L, Veverkova L, Vlcek P, Katolicka J, Capov I, Hermanova M, Smarda J, et al. Overexpression of c-Myb is associated with suppression of distant metastases in colorectal carcinoma. Tumour Biol. 2016;37(8):10723–9.PubMedCrossRef
39.
go back to reference Tichy M, Knopfova L, Jarkovsky J, Vlcek P, Katolicka J, Capov I, Hermanova M, Smarda J, Benes P. High c-Myb expression associates with good prognosis in colorectal carcinoma. J Cancer. 2019;10(6):1393–7.PubMedPubMedCentralCrossRef Tichy M, Knopfova L, Jarkovsky J, Vlcek P, Katolicka J, Capov I, Hermanova M, Smarda J, Benes P. High c-Myb expression associates with good prognosis in colorectal carcinoma. J Cancer. 2019;10(6):1393–7.PubMedPubMedCentralCrossRef
40.
go back to reference Millen R, Malaterre J, Cross RS, Carpinteri S, Desai J, Tran B, Darcy P, Gibbs P, Sieber O, Zeps N, et al. Immunomodulation by MYB is associated with tumor relapse in patients with early stage colorectal cancer. Oncoimmunology. 2016;5(7):e1149667.PubMedPubMedCentralCrossRef Millen R, Malaterre J, Cross RS, Carpinteri S, Desai J, Tran B, Darcy P, Gibbs P, Sieber O, Zeps N, et al. Immunomodulation by MYB is associated with tumor relapse in patients with early stage colorectal cancer. Oncoimmunology. 2016;5(7):e1149667.PubMedPubMedCentralCrossRef
41.
go back to reference Olsen J, Espersen ML, Jess P, Kirkeby LT, Troelsen JT. The clinical perspectives of CDX2 expression in colorectal cancer: a qualitative systematic review. Surg Oncol. 2014;23(3):167–76.PubMedCrossRef Olsen J, Espersen ML, Jess P, Kirkeby LT, Troelsen JT. The clinical perspectives of CDX2 expression in colorectal cancer: a qualitative systematic review. Surg Oncol. 2014;23(3):167–76.PubMedCrossRef
42.
go back to reference Sandberg TP, Sweere I, van Pelt GW, Putter H, Vermeulen L, Kuppen PJ, Tollenaar R, Mesker WE. Prognostic value of low CDX2 expression in colorectal cancers with a high stromal content—a short report. Cell Oncol (Dordr). 2019;42(3):397–403.CrossRef Sandberg TP, Sweere I, van Pelt GW, Putter H, Vermeulen L, Kuppen PJ, Tollenaar R, Mesker WE. Prognostic value of low CDX2 expression in colorectal cancers with a high stromal content—a short report. Cell Oncol (Dordr). 2019;42(3):397–403.CrossRef
43.
go back to reference Zanella ER, Galimi F, Sassi F, Migliardi G, Cottino F, Leto SM, Lupo B, Erriquez J, Isella C, Comoglio PM, et al. IGF2 is an actionable target that identifies a distinct subpopulation of colorectal cancer patients with marginal response to anti-EGFR therapies. Sci Transl Med. 2015;7(272):272ra212.CrossRef Zanella ER, Galimi F, Sassi F, Migliardi G, Cottino F, Leto SM, Lupo B, Erriquez J, Isella C, Comoglio PM, et al. IGF2 is an actionable target that identifies a distinct subpopulation of colorectal cancer patients with marginal response to anti-EGFR therapies. Sci Transl Med. 2015;7(272):272ra212.CrossRef
44.
go back to reference Tauriello DVF, Palomo-Ponce S, Stork D, Berenguer-Llergo A, Badia-Ramentol J, Iglesias M, Sevillano M, Ibiza S, Canellas A, Hernando-Momblona X, et al. TGFbeta drives immune evasion in genetically reconstituted colon cancer metastasis. Nature. 2018;554(7693):538–43.PubMedCrossRef Tauriello DVF, Palomo-Ponce S, Stork D, Berenguer-Llergo A, Badia-Ramentol J, Iglesias M, Sevillano M, Ibiza S, Canellas A, Hernando-Momblona X, et al. TGFbeta drives immune evasion in genetically reconstituted colon cancer metastasis. Nature. 2018;554(7693):538–43.PubMedCrossRef
45.
go back to reference Boland PM, Ma WW. Immunotherapy for colorectal cancer. Cancers (Basel). 2017;9(5):50.CrossRef Boland PM, Ma WW. Immunotherapy for colorectal cancer. Cancers (Basel). 2017;9(5):50.CrossRef
46.
go back to reference Le DT, Kim TW, Van Cutsem E, Geva R, Jager D, Hara H, Burge M, O’Neil B, Kavan P, Yoshino T, et al. Phase II open-label study of pembrolizumab in treatment-refractory, microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: KEYNOTE-164. J Clin Oncol. 2020;38(1):11–9.PubMedCrossRef Le DT, Kim TW, Van Cutsem E, Geva R, Jager D, Hara H, Burge M, O’Neil B, Kavan P, Yoshino T, et al. Phase II open-label study of pembrolizumab in treatment-refractory, microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: KEYNOTE-164. J Clin Oncol. 2020;38(1):11–9.PubMedCrossRef
47.
go back to reference Inoue Y, Hazama S, Suzuki N, Tokumitsu Y, Kanekiyo S, Tomochika S, Tsunedomi R, Tokuhisa Y, Iida M, Sakamoto K, et al. Cetuximab strongly enhances immune cell infiltration into liver metastatic sites in colorectal cancer. Cancer Sci. 2017;108(3):455–60.PubMedPubMedCentralCrossRef Inoue Y, Hazama S, Suzuki N, Tokumitsu Y, Kanekiyo S, Tomochika S, Tsunedomi R, Tokuhisa Y, Iida M, Sakamoto K, et al. Cetuximab strongly enhances immune cell infiltration into liver metastatic sites in colorectal cancer. Cancer Sci. 2017;108(3):455–60.PubMedPubMedCentralCrossRef
48.
go back to reference Wang L, Wei Y, Fang W, Lu C, Chen J, Cui G, Diao H. Cetuximab enhanced the cytotoxic activity of immune cells during treatment of colorectal cancer. Cell Physiol Biochem. 2017;44(3):1038–50.PubMedCrossRef Wang L, Wei Y, Fang W, Lu C, Chen J, Cui G, Diao H. Cetuximab enhanced the cytotoxic activity of immune cells during treatment of colorectal cancer. Cell Physiol Biochem. 2017;44(3):1038–50.PubMedCrossRef
49.
go back to reference Sunakawa Y, Yang D, Cao S, Zhang W, Moran M, Astrow SH, Hsiang J, Stephens C, Tsuji A, Takahashi T, et al. Immune-related genes to dominate Neutrophil-lymphocyte Ratio (NLR) associated with survival of cetuximab treatment in metastatic colorectal cancer. Clin Colorectal Cancer. 2018;17(4):e741-9.PubMedPubMedCentralCrossRef Sunakawa Y, Yang D, Cao S, Zhang W, Moran M, Astrow SH, Hsiang J, Stephens C, Tsuji A, Takahashi T, et al. Immune-related genes to dominate Neutrophil-lymphocyte Ratio (NLR) associated with survival of cetuximab treatment in metastatic colorectal cancer. Clin Colorectal Cancer. 2018;17(4):e741-9.PubMedPubMedCentralCrossRef
Metadata
Title
Identification of key genes involved in tumor immune cell infiltration and cetuximab resistance in colorectal cancer
Authors
Li Liang
Mengling Liu
Xun Sun
Yitao Yuan
Ke Peng
Khalid Rashid
Yiyi Yu
Yuehong Cui
Yanjie Chen
Tianshu Liu
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/s12935-021-01829-8

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