Skip to main content
Top
Published in: Journal of Translational Medicine 1/2018

Open Access 01-12-2018 | Research

In silico identification of drug target pathways in breast cancer subtypes using pathway cross-talk inhibition

Authors: Claudia Cava, Gloria Bertoli, Isabella Castiglioni

Published in: Journal of Translational Medicine | Issue 1/2018

Login to get access

Abstract

Background

Despite great development in genome and proteome high-throughput methods, treatment failure is a critical point in the management of most solid cancers, including breast cancer (BC). Multiple alternative mechanisms upon drug treatment are involved to offset therapeutic effects, eventually causing drug resistance or treatment failure.

Methods

Here, we optimized a computational method to discover novel drug target pathways in cancer subtypes using pathway cross-talk inhibition (PCI). The in silico method is based on the detection and quantification of the pathway cross-talk for distinct cancer subtypes. From a BC data set of The Cancer Genome Atlas, we have identified different networks of cross-talking pathways for different BC subtypes, validated using an independent BC dataset from Gene Expression Omnibus. Then, we predicted in silico the effects of new or approved drugs on different BC subtypes by silencing individual or combined subtype-derived pathways with the aim to find new potential drugs or more effective synergistic combinations of drugs.

Results

Overall, we identified a set of new potential drug target pathways for distinct BC subtypes on which therapeutic agents could synergically act showing antitumour effects and impacting on cross-talk inhibition.

Conclusions

We believe that in silico methods based on PCI could offer valuable approaches to identifying more tailored and effective treatments in particular in heterogeneous cancer diseases.
Appendix
Available only for authorised users
Literature
2.
go back to reference Csermely P, Agoston V, Pongor S. The efficiency of multi-target drugs: the network approach might help drug design. Trends Pharmacol Sci. 2005;26(4):178–82.CrossRefPubMed Csermely P, Agoston V, Pongor S. The efficiency of multi-target drugs: the network approach might help drug design. Trends Pharmacol Sci. 2005;26(4):178–82.CrossRefPubMed
4.
go back to reference Duncan JS, Whittle MC, Nakamura K, Abell AN, Midland AA, Zawistowski JS, Johnson NL, Granger DA, Jordan NV, Darr DB, Usary J, Kuan PF, Smalley DM, Major B, He X, Hoadley KA, Zhou B, Sharpless NE, Perou CM, Kim WY, Gomez SM, Chen X, Jin J, Frye SV, Earp HS, Graves LM, Johnson GL. Dynamic reprogramming of the kinome in response to targeted MEK inhibition in triple-negative breast cancer. Cell. 2012;149(2):307–21. https://doi.org/10.1016/j.cell.2012.02.053.CrossRefPubMedPubMedCentral Duncan JS, Whittle MC, Nakamura K, Abell AN, Midland AA, Zawistowski JS, Johnson NL, Granger DA, Jordan NV, Darr DB, Usary J, Kuan PF, Smalley DM, Major B, He X, Hoadley KA, Zhou B, Sharpless NE, Perou CM, Kim WY, Gomez SM, Chen X, Jin J, Frye SV, Earp HS, Graves LM, Johnson GL. Dynamic reprogramming of the kinome in response to targeted MEK inhibition in triple-negative breast cancer. Cell. 2012;149(2):307–21. https://​doi.​org/​10.​1016/​j.​cell.​2012.​02.​053.CrossRefPubMedPubMedCentral
5.
go back to reference Yamaguchi H, Chang SS, Hsu JL, Hung MC. Signaling cross-talk in the resistance to HER family receptor targeted therapy. Oncogene. 2014;33(9):1073–81.CrossRefPubMed Yamaguchi H, Chang SS, Hsu JL, Hung MC. Signaling cross-talk in the resistance to HER family receptor targeted therapy. Oncogene. 2014;33(9):1073–81.CrossRefPubMed
9.
go back to reference van de Vijver MJ, He YD, van’t Veer LJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002;347:1999–2009.CrossRefPubMed van de Vijver MJ, He YD, van’t Veer LJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002;347:1999–2009.CrossRefPubMed
10.
go back to reference Günther S, Kuhn M, Dunkel M, Campillos M, Senger C, Petsalaki E, Ahmed J, Urdiales EG, Gewiess A, Jensen LJ, Schneider R, Skoblo R, Russell RB, Bourne PE, Bork P, Preissner R. SuperTarget and Matador: resources for exploring drug-target relationships. Nucleic Acids Res. 2008;36(Database issue):D919–22.PubMed Günther S, Kuhn M, Dunkel M, Campillos M, Senger C, Petsalaki E, Ahmed J, Urdiales EG, Gewiess A, Jensen LJ, Schneider R, Skoblo R, Russell RB, Bourne PE, Bork P, Preissner R. SuperTarget and Matador: resources for exploring drug-target relationships. Nucleic Acids Res. 2008;36(Database issue):D919–22.PubMed
12.
14.
go back to reference Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995;57(1):289–300. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995;57(1):289–300.
15.
go back to reference Graudenzi A, Cava C, Bertoli G, Fromm B, Flatmark K, Mauri G, Castiglioni I. Pathway-based classification of breast cancer subtypes. Front Biosci (Landmark Ed). 2017;22:1697–712.CrossRefPubMed Graudenzi A, Cava C, Bertoli G, Fromm B, Flatmark K, Mauri G, Castiglioni I. Pathway-based classification of breast cancer subtypes. Front Biosci (Landmark Ed). 2017;22:1697–712.CrossRefPubMed
16.
go back to reference Liaw A, Wiener M. Classification and regression by randomforest. R News. 2002;2(3):18–22. Liaw A, Wiener M. Classification and regression by randomforest. R News. 2002;2(3):18–22.
18.
go back to reference Cascante M, Boros LG, Comin-Anduix B, de Atauri P, Centelles JJ, Lee PW. Metabolic control analysis in drug discovery and disease. Nat Biotechnol. 2002;20(3):243–9.CrossRefPubMed Cascante M, Boros LG, Comin-Anduix B, de Atauri P, Centelles JJ, Lee PW. Metabolic control analysis in drug discovery and disease. Nat Biotechnol. 2002;20(3):243–9.CrossRefPubMed
19.
20.
go back to reference Archakov AI, Govorun VM, Dubanov AV, Ivanov YD, Veselovsky AV, Lewi P, Janssen P. Protein–protein interactions as a target for drugs in proteomics. Proteomics. 2003;3(4):380–91.CrossRefPubMed Archakov AI, Govorun VM, Dubanov AV, Ivanov YD, Veselovsky AV, Lewi P, Janssen P. Protein–protein interactions as a target for drugs in proteomics. Proteomics. 2003;3(4):380–91.CrossRefPubMed
21.
go back to reference Kazmirchuk T, Dick K, Burnside DJ, Barnes B, Moteshareie H, Hajikarimlou M, Omidi K, Ahmed D, Low A, Lettl C, Hooshyar M, Schoenrock A, Pitre S, Babu M, Cassol E, Samanfar B, Wong A, Dehne F, Green JR, Golshani A. Designing anti-Zika virus peptides derived from predicted human-Zika virus protein–protein interactions. Comput Biol Chem. 2017;71:180–7.CrossRefPubMed Kazmirchuk T, Dick K, Burnside DJ, Barnes B, Moteshareie H, Hajikarimlou M, Omidi K, Ahmed D, Low A, Lettl C, Hooshyar M, Schoenrock A, Pitre S, Babu M, Cassol E, Samanfar B, Wong A, Dehne F, Green JR, Golshani A. Designing anti-Zika virus peptides derived from predicted human-Zika virus protein–protein interactions. Comput Biol Chem. 2017;71:180–7.CrossRefPubMed
23.
go back to reference Ali S, Coombes RC. Estrogen receptor alpha in human breast cancer: occurrence and significance. J Mammary Gland Biol Neoplasia. 2000;5(3):271–81.CrossRefPubMed Ali S, Coombes RC. Estrogen receptor alpha in human breast cancer: occurrence and significance. J Mammary Gland Biol Neoplasia. 2000;5(3):271–81.CrossRefPubMed
24.
go back to reference Fukuchi J, Kokontis JM, Hiipakka RA, Chuu CP, Liao S. Antiproliferative effect of liver X receptor agonists on LNCaP human prostate cancer cells. Cancer Res. 2004;64(21):7686–9.CrossRefPubMed Fukuchi J, Kokontis JM, Hiipakka RA, Chuu CP, Liao S. Antiproliferative effect of liver X receptor agonists on LNCaP human prostate cancer cells. Cancer Res. 2004;64(21):7686–9.CrossRefPubMed
27.
go back to reference El Roz A, Bard JM, Huvelin JM, Nazih H. LXR agonists and ABCG1-dependent cholesterol efflux in MCF-7 breast cancer cells: relation to proliferation and apoptosis. Anticancer Res. 2012;32(7):3007–13.PubMed El Roz A, Bard JM, Huvelin JM, Nazih H. LXR agonists and ABCG1-dependent cholesterol efflux in MCF-7 breast cancer cells: relation to proliferation and apoptosis. Anticancer Res. 2012;32(7):3007–13.PubMed
28.
go back to reference Gong H, Guo P, Zhai Y, Zhou J, Uppal H, Jarzynka MJ, Song WC, Cheng SY, Xie W. Estrogen deprivation and inhibition of breast cancer growth in vivo through activation of the orphan nuclear receptor liver X receptor. Mol Endocrinol. 2007;21(8):1781–90.CrossRefPubMed Gong H, Guo P, Zhai Y, Zhou J, Uppal H, Jarzynka MJ, Song WC, Cheng SY, Xie W. Estrogen deprivation and inhibition of breast cancer growth in vivo through activation of the orphan nuclear receptor liver X receptor. Mol Endocrinol. 2007;21(8):1781–90.CrossRefPubMed
30.
go back to reference Ferlini C, Scambia G, Distefano M, Filippini P, Isola G, Riva A, Bombardelli E, Fattorossi A, Benedetti Panici P, Mancuso S. Synergistic antiproliferative activity of tamoxifen and docetaxel on three oestrogen receptor-negative cancer cell lines is mediated by the induction of apoptosis. Br J Cancer. 1997;75(6):884–91.CrossRefPubMedPubMedCentral Ferlini C, Scambia G, Distefano M, Filippini P, Isola G, Riva A, Bombardelli E, Fattorossi A, Benedetti Panici P, Mancuso S. Synergistic antiproliferative activity of tamoxifen and docetaxel on three oestrogen receptor-negative cancer cell lines is mediated by the induction of apoptosis. Br J Cancer. 1997;75(6):884–91.CrossRefPubMedPubMedCentral
Metadata
Title
In silico identification of drug target pathways in breast cancer subtypes using pathway cross-talk inhibition
Authors
Claudia Cava
Gloria Bertoli
Isabella Castiglioni
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Translational Medicine / Issue 1/2018
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-018-1535-2

Other articles of this Issue 1/2018

Journal of Translational Medicine 1/2018 Go to the issue