Home > Research > Publications & Outputs > Decitabine Response in Breast Cancer Requires E...
View graph of relations

Decitabine Response in Breast Cancer Requires Efficient Drug Processing and Is Not Limited by Multidrug Resistance

Research output: Contribution to Journal/MagazineJournal articlepeer-review

Published

Standard

Decitabine Response in Breast Cancer Requires Efficient Drug Processing and Is Not Limited by Multidrug Resistance. / Dahn, Margaret L. ; Cruickshank, Brianne M.; Jackson, Ainsleigh J. et al.
In: Molecular Cancer Therapeutics, Vol. 19, No. 5, 04.05.2020, p. 1110–1122.

Research output: Contribution to Journal/MagazineJournal articlepeer-review

Harvard

Dahn, ML, Cruickshank, BM, Jackson, AJ, Dean, C, Holloway, RW, Hall, S, Coyle, KM, Maillet, H, Waisman, DM, Goralski, KB, Giacomantonio, CA, Weaver, ICG & Marcato, P 2020, 'Decitabine Response in Breast Cancer Requires Efficient Drug Processing and Is Not Limited by Multidrug Resistance', Molecular Cancer Therapeutics, vol. 19, no. 5, pp. 1110–1122. https://doi.org/10.1158/1535-7163.MCT-19-0745

APA

Dahn, M. L., Cruickshank, B. M., Jackson, A. J., Dean, C., Holloway, R. W., Hall, S., Coyle, K. M., Maillet, H., Waisman, D. M., Goralski, K. B., Giacomantonio, C. A., Weaver, I. C. G., & Marcato, P. (2020). Decitabine Response in Breast Cancer Requires Efficient Drug Processing and Is Not Limited by Multidrug Resistance. Molecular Cancer Therapeutics, 19(5), 1110–1122. https://doi.org/10.1158/1535-7163.MCT-19-0745

Vancouver

Dahn ML, Cruickshank BM, Jackson AJ, Dean C, Holloway RW, Hall S et al. Decitabine Response in Breast Cancer Requires Efficient Drug Processing and Is Not Limited by Multidrug Resistance. Molecular Cancer Therapeutics. 2020 May 4;19(5):1110–1122. doi: 10.1158/1535-7163.MCT-19-0745

Author

Dahn, Margaret L. ; Cruickshank, Brianne M. ; Jackson, Ainsleigh J. et al. / Decitabine Response in Breast Cancer Requires Efficient Drug Processing and Is Not Limited by Multidrug Resistance. In: Molecular Cancer Therapeutics. 2020 ; Vol. 19, No. 5. pp. 1110–1122.

Bibtex

@article{998cfb1e275d4cdd84abef4db7af2037,
title = "Decitabine Response in Breast Cancer Requires Efficient Drug Processing and Is Not Limited by Multidrug Resistance",
abstract = "Dysregulation of DNA methylation is an established feature of breast cancers. DNA demethylating therapies like decitabine are proposed for the treatment of triple-negative breast cancers (TNBC) and indicators of response need to be identified. For this purpose, we characterized the effects of decitabine in a panel of 10 breast cancer cell lines and observed a range of sensitivity to decitabine that was not subtype specific. Knockdown of potential key effectors demonstrated the requirement of deoxycytidine kinase (DCK) for decitabine response in breast cancer cells. In treatment-na{\"i}ve breast tumors, DCK was higher in TNBCs, and DCK levels were sustained or increased post chemotherapy treatment. This suggests that limited DCK levels will not be a barrier to response in patients with TNBC treated with decitabine as a second-line treatment or in a clinical trial. Methylome analysis revealed that genome-wide, region-specific, tumor suppressor gene–specific methylation, and decitabine-induced demethylation did not predict response to decitabine. Gene set enrichment analysis of transcriptome data demonstrated that decitabine induced genes within apoptosis, cell cycle, stress, and immune pathways. Induced genes included those characterized by the viral mimicry response; however, knockdown of key effectors of the pathway did not affect decitabine sensitivity suggesting that breast cancer growth suppression by decitabine is independent of viral mimicry. Finally, taxol-resistant breast cancer cells expressing high levels of multidrug resistance transporter ABCB1 remained sensitive to decitabine, suggesting that the drug could be used as second-line treatment for chemoresistant patients.",
author = "Dahn, {Margaret L.} and Cruickshank, {Brianne M.} and Jackson, {Ainsleigh J.} and Cheryl Dean and Holloway, {Ryan W.} and Steve Hall and Coyle, {Krysta M.} and Hillary Maillet and Waisman, {David M.} and Goralski, {Kerry B} and Giacomantonio, {Carman A.} and Weaver, {Ian C. G.} and Paola Marcato",
year = "2020",
month = may,
day = "4",
doi = "10.1158/1535-7163.MCT-19-0745",
language = "English",
volume = "19",
pages = "1110–1122",
journal = "Molecular Cancer Therapeutics",
number = "5",

}

RIS

TY - JOUR

T1 - Decitabine Response in Breast Cancer Requires Efficient Drug Processing and Is Not Limited by Multidrug Resistance

AU - Dahn, Margaret L.

AU - Cruickshank, Brianne M.

AU - Jackson, Ainsleigh J.

AU - Dean, Cheryl

AU - Holloway, Ryan W.

AU - Hall, Steve

AU - Coyle, Krysta M.

AU - Maillet, Hillary

AU - Waisman, David M.

AU - Goralski, Kerry B

AU - Giacomantonio, Carman A.

AU - Weaver, Ian C. G.

AU - Marcato, Paola

PY - 2020/5/4

Y1 - 2020/5/4

N2 - Dysregulation of DNA methylation is an established feature of breast cancers. DNA demethylating therapies like decitabine are proposed for the treatment of triple-negative breast cancers (TNBC) and indicators of response need to be identified. For this purpose, we characterized the effects of decitabine in a panel of 10 breast cancer cell lines and observed a range of sensitivity to decitabine that was not subtype specific. Knockdown of potential key effectors demonstrated the requirement of deoxycytidine kinase (DCK) for decitabine response in breast cancer cells. In treatment-naïve breast tumors, DCK was higher in TNBCs, and DCK levels were sustained or increased post chemotherapy treatment. This suggests that limited DCK levels will not be a barrier to response in patients with TNBC treated with decitabine as a second-line treatment or in a clinical trial. Methylome analysis revealed that genome-wide, region-specific, tumor suppressor gene–specific methylation, and decitabine-induced demethylation did not predict response to decitabine. Gene set enrichment analysis of transcriptome data demonstrated that decitabine induced genes within apoptosis, cell cycle, stress, and immune pathways. Induced genes included those characterized by the viral mimicry response; however, knockdown of key effectors of the pathway did not affect decitabine sensitivity suggesting that breast cancer growth suppression by decitabine is independent of viral mimicry. Finally, taxol-resistant breast cancer cells expressing high levels of multidrug resistance transporter ABCB1 remained sensitive to decitabine, suggesting that the drug could be used as second-line treatment for chemoresistant patients.

AB - Dysregulation of DNA methylation is an established feature of breast cancers. DNA demethylating therapies like decitabine are proposed for the treatment of triple-negative breast cancers (TNBC) and indicators of response need to be identified. For this purpose, we characterized the effects of decitabine in a panel of 10 breast cancer cell lines and observed a range of sensitivity to decitabine that was not subtype specific. Knockdown of potential key effectors demonstrated the requirement of deoxycytidine kinase (DCK) for decitabine response in breast cancer cells. In treatment-naïve breast tumors, DCK was higher in TNBCs, and DCK levels were sustained or increased post chemotherapy treatment. This suggests that limited DCK levels will not be a barrier to response in patients with TNBC treated with decitabine as a second-line treatment or in a clinical trial. Methylome analysis revealed that genome-wide, region-specific, tumor suppressor gene–specific methylation, and decitabine-induced demethylation did not predict response to decitabine. Gene set enrichment analysis of transcriptome data demonstrated that decitabine induced genes within apoptosis, cell cycle, stress, and immune pathways. Induced genes included those characterized by the viral mimicry response; however, knockdown of key effectors of the pathway did not affect decitabine sensitivity suggesting that breast cancer growth suppression by decitabine is independent of viral mimicry. Finally, taxol-resistant breast cancer cells expressing high levels of multidrug resistance transporter ABCB1 remained sensitive to decitabine, suggesting that the drug could be used as second-line treatment for chemoresistant patients.

U2 - 10.1158/1535-7163.MCT-19-0745

DO - 10.1158/1535-7163.MCT-19-0745

M3 - Journal article

VL - 19

SP - 1110

EP - 1122

JO - Molecular Cancer Therapeutics

JF - Molecular Cancer Therapeutics

IS - 5

ER -