Identification of selective cytotoxic and synthetic lethal drug responses in triple negative breast cancer cells

Research output: Contribution to journalJournal articleResearchpeer-review

Standard

Identification of selective cytotoxic and synthetic lethal drug responses in triple negative breast cancer cells. / Gautam, Prson; Karhinen, Leena; Szwajda, Agnieszka; Jha, Sawan Kumar; Yadav, Bhagwan; Aittokallio, Tero; Wennerberg, Krister.

In: Molecular Cancer, Vol. 15, No. 1, 10.05.2016, p. 34.

Research output: Contribution to journalJournal articleResearchpeer-review

Harvard

Gautam, P, Karhinen, L, Szwajda, A, Jha, SK, Yadav, B, Aittokallio, T & Wennerberg, K 2016, 'Identification of selective cytotoxic and synthetic lethal drug responses in triple negative breast cancer cells', Molecular Cancer, vol. 15, no. 1, pp. 34. https://doi.org/10.1186/s12943-016-0517-3

APA

Gautam, P., Karhinen, L., Szwajda, A., Jha, S. K., Yadav, B., Aittokallio, T., & Wennerberg, K. (2016). Identification of selective cytotoxic and synthetic lethal drug responses in triple negative breast cancer cells. Molecular Cancer, 15(1), 34. https://doi.org/10.1186/s12943-016-0517-3

Vancouver

Gautam P, Karhinen L, Szwajda A, Jha SK, Yadav B, Aittokallio T et al. Identification of selective cytotoxic and synthetic lethal drug responses in triple negative breast cancer cells. Molecular Cancer. 2016 May 10;15(1):34. https://doi.org/10.1186/s12943-016-0517-3

Author

Gautam, Prson ; Karhinen, Leena ; Szwajda, Agnieszka ; Jha, Sawan Kumar ; Yadav, Bhagwan ; Aittokallio, Tero ; Wennerberg, Krister. / Identification of selective cytotoxic and synthetic lethal drug responses in triple negative breast cancer cells. In: Molecular Cancer. 2016 ; Vol. 15, No. 1. pp. 34.

Bibtex

@article{2419f0fbd35b45e79535b3b64fcad600,
title = "Identification of selective cytotoxic and synthetic lethal drug responses in triple negative breast cancer cells",
abstract = "BACKGROUND: Triple negative breast cancer (TNBC) is a highly heterogeneous and aggressive type of cancer that lacks effective targeted therapy. Despite detailed molecular profiling, no targeted therapy has been established. Hence, with the aim of gaining deeper understanding of the functional differences of TNBC subtypes and how that may relate to potential novel therapeutic strategies, we studied comprehensive anticancer-agent responses among a panel of TNBC cell lines.METHOD: The responses of 301 approved and investigational oncology compounds were measured in 16 TNBC cell lines applying a functional profiling approach. To go beyond the standard drug viability effect profiling, which has been used in most chemosensitivity studies, we utilized a multiplexed readout for both cell viability and cytotoxicity, allowing us to differentiate between cytostatic and cytotoxic responses.RESULTS: Our approach revealed that most single-agent anti-cancer compounds that showed activity for the viability readout had no or little cytotoxic effects. Major compound classes that exhibited this type of response included anti-mitotics, mTOR, CDK, and metabolic inhibitors, as well as many agents selectively inhibiting oncogene-activated pathways. However, within the broad viability-acting classes of compounds, there were often subsets of cell lines that responded by cell death, suggesting that these cells are particularly vulnerable to the tested substance. In those cases we could identify differential levels of protein markers associated with cytotoxic responses. For example, PAI-1, MAPK phosphatase and Notch-3 levels associated with cytotoxic responses to mitotic and proteasome inhibitors, suggesting that these might serve as markers of response also in clinical settings. Furthermore, the cytotoxicity readout highlighted selective synergistic and synthetic lethal drug combinations that were missed by the cell viability readouts. For instance, the MEK inhibitor trametinib synergized with PARP inhibitors. Similarly, combination of two non-cytotoxic compounds, the rapamycin analog everolimus and an ATP-competitive mTOR inhibitor dactolisib, showed synthetic lethality in several mTOR-addicted cell lines.CONCLUSIONS: Taken together, by studying the combination of cytotoxic and cytostatic drug responses, we identified a deeper spectrum of cellular responses both to single agents and combinations that may be highly relevant for identifying precision medicine approaches in TNBC as well as in other types of cancers.",
keywords = "Antineoplastic Agents/pharmacology, Biomarkers, Biomarkers, Tumor, Cell Cycle Checkpoints/drug effects, Cell Line, Tumor, Cell Survival/drug effects, Cluster Analysis, Computational Biology, Drug Discovery, Drug Resistance, Neoplasm/genetics, Drug Screening Assays, Antitumor, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic/drug effects, Humans, Protein Kinase Inhibitors/pharmacology, Synthetic Lethal Mutations/drug effects, TOR Serine-Threonine Kinases/antagonists & inhibitors, Transcriptome, Triple Negative Breast Neoplasms/drug therapy",
author = "Prson Gautam and Leena Karhinen and Agnieszka Szwajda and Jha, {Sawan Kumar} and Bhagwan Yadav and Tero Aittokallio and Krister Wennerberg",
year = "2016",
month = may,
day = "10",
doi = "10.1186/s12943-016-0517-3",
language = "English",
volume = "15",
pages = "34",
journal = "Molecular Cancer",
issn = "1476-4598",
publisher = "BioMed Central",
number = "1",

}

RIS

TY - JOUR

T1 - Identification of selective cytotoxic and synthetic lethal drug responses in triple negative breast cancer cells

AU - Gautam, Prson

AU - Karhinen, Leena

AU - Szwajda, Agnieszka

AU - Jha, Sawan Kumar

AU - Yadav, Bhagwan

AU - Aittokallio, Tero

AU - Wennerberg, Krister

PY - 2016/5/10

Y1 - 2016/5/10

N2 - BACKGROUND: Triple negative breast cancer (TNBC) is a highly heterogeneous and aggressive type of cancer that lacks effective targeted therapy. Despite detailed molecular profiling, no targeted therapy has been established. Hence, with the aim of gaining deeper understanding of the functional differences of TNBC subtypes and how that may relate to potential novel therapeutic strategies, we studied comprehensive anticancer-agent responses among a panel of TNBC cell lines.METHOD: The responses of 301 approved and investigational oncology compounds were measured in 16 TNBC cell lines applying a functional profiling approach. To go beyond the standard drug viability effect profiling, which has been used in most chemosensitivity studies, we utilized a multiplexed readout for both cell viability and cytotoxicity, allowing us to differentiate between cytostatic and cytotoxic responses.RESULTS: Our approach revealed that most single-agent anti-cancer compounds that showed activity for the viability readout had no or little cytotoxic effects. Major compound classes that exhibited this type of response included anti-mitotics, mTOR, CDK, and metabolic inhibitors, as well as many agents selectively inhibiting oncogene-activated pathways. However, within the broad viability-acting classes of compounds, there were often subsets of cell lines that responded by cell death, suggesting that these cells are particularly vulnerable to the tested substance. In those cases we could identify differential levels of protein markers associated with cytotoxic responses. For example, PAI-1, MAPK phosphatase and Notch-3 levels associated with cytotoxic responses to mitotic and proteasome inhibitors, suggesting that these might serve as markers of response also in clinical settings. Furthermore, the cytotoxicity readout highlighted selective synergistic and synthetic lethal drug combinations that were missed by the cell viability readouts. For instance, the MEK inhibitor trametinib synergized with PARP inhibitors. Similarly, combination of two non-cytotoxic compounds, the rapamycin analog everolimus and an ATP-competitive mTOR inhibitor dactolisib, showed synthetic lethality in several mTOR-addicted cell lines.CONCLUSIONS: Taken together, by studying the combination of cytotoxic and cytostatic drug responses, we identified a deeper spectrum of cellular responses both to single agents and combinations that may be highly relevant for identifying precision medicine approaches in TNBC as well as in other types of cancers.

AB - BACKGROUND: Triple negative breast cancer (TNBC) is a highly heterogeneous and aggressive type of cancer that lacks effective targeted therapy. Despite detailed molecular profiling, no targeted therapy has been established. Hence, with the aim of gaining deeper understanding of the functional differences of TNBC subtypes and how that may relate to potential novel therapeutic strategies, we studied comprehensive anticancer-agent responses among a panel of TNBC cell lines.METHOD: The responses of 301 approved and investigational oncology compounds were measured in 16 TNBC cell lines applying a functional profiling approach. To go beyond the standard drug viability effect profiling, which has been used in most chemosensitivity studies, we utilized a multiplexed readout for both cell viability and cytotoxicity, allowing us to differentiate between cytostatic and cytotoxic responses.RESULTS: Our approach revealed that most single-agent anti-cancer compounds that showed activity for the viability readout had no or little cytotoxic effects. Major compound classes that exhibited this type of response included anti-mitotics, mTOR, CDK, and metabolic inhibitors, as well as many agents selectively inhibiting oncogene-activated pathways. However, within the broad viability-acting classes of compounds, there were often subsets of cell lines that responded by cell death, suggesting that these cells are particularly vulnerable to the tested substance. In those cases we could identify differential levels of protein markers associated with cytotoxic responses. For example, PAI-1, MAPK phosphatase and Notch-3 levels associated with cytotoxic responses to mitotic and proteasome inhibitors, suggesting that these might serve as markers of response also in clinical settings. Furthermore, the cytotoxicity readout highlighted selective synergistic and synthetic lethal drug combinations that were missed by the cell viability readouts. For instance, the MEK inhibitor trametinib synergized with PARP inhibitors. Similarly, combination of two non-cytotoxic compounds, the rapamycin analog everolimus and an ATP-competitive mTOR inhibitor dactolisib, showed synthetic lethality in several mTOR-addicted cell lines.CONCLUSIONS: Taken together, by studying the combination of cytotoxic and cytostatic drug responses, we identified a deeper spectrum of cellular responses both to single agents and combinations that may be highly relevant for identifying precision medicine approaches in TNBC as well as in other types of cancers.

KW - Antineoplastic Agents/pharmacology

KW - Biomarkers

KW - Biomarkers, Tumor

KW - Cell Cycle Checkpoints/drug effects

KW - Cell Line, Tumor

KW - Cell Survival/drug effects

KW - Cluster Analysis

KW - Computational Biology

KW - Drug Discovery

KW - Drug Resistance, Neoplasm/genetics

KW - Drug Screening Assays, Antitumor

KW - Female

KW - Gene Expression Profiling

KW - Gene Expression Regulation, Neoplastic/drug effects

KW - Humans

KW - Protein Kinase Inhibitors/pharmacology

KW - Synthetic Lethal Mutations/drug effects

KW - TOR Serine-Threonine Kinases/antagonists & inhibitors

KW - Transcriptome

KW - Triple Negative Breast Neoplasms/drug therapy

U2 - 10.1186/s12943-016-0517-3

DO - 10.1186/s12943-016-0517-3

M3 - Journal article

C2 - 27165605

VL - 15

SP - 34

JO - Molecular Cancer

JF - Molecular Cancer

SN - 1476-4598

IS - 1

ER -

ID: 199424289