Wee1 Inhibition Enhances the Anti-Tumor Effects of Capecitabine in Preclinical Models of Triple-Negative Breast Cancer

Triple-negative breast cancer (TNBC) is an aggressive subtype defined by lack of hormone receptor expression and non-amplified HER2. Adavosertib (AZD1775) is a potent, small-molecule, ATP-competitive inhibitor of the Wee1 kinase that potentiates the activity of many DNA-damaging chemotherapeutics an...

Full description

Bibliographic Details
Main Authors: Todd M. Pitts, Dennis M. Simmons, Stacey M. Bagby, Sarah J. Hartman, Betelehem W. Yacob, Brian Gittleman, John J. Tentler, Diana Cittelly, D. Ryan Ormond, Wells A. Messersmith, S. Gail Eckhardt, Jennifer R. Diamond
Format: Article
Language:English
Published: MDPI AG 2020-03-01
Series:Cancers
Subjects:
Online Access:https://www.mdpi.com/2072-6694/12/3/719
_version_ 1797764666475151360
author Todd M. Pitts
Dennis M. Simmons
Stacey M. Bagby
Sarah J. Hartman
Betelehem W. Yacob
Brian Gittleman
John J. Tentler
Diana Cittelly
D. Ryan Ormond
Wells A. Messersmith
S. Gail Eckhardt
Jennifer R. Diamond
author_facet Todd M. Pitts
Dennis M. Simmons
Stacey M. Bagby
Sarah J. Hartman
Betelehem W. Yacob
Brian Gittleman
John J. Tentler
Diana Cittelly
D. Ryan Ormond
Wells A. Messersmith
S. Gail Eckhardt
Jennifer R. Diamond
author_sort Todd M. Pitts
collection DOAJ
description Triple-negative breast cancer (TNBC) is an aggressive subtype defined by lack of hormone receptor expression and non-amplified HER2. Adavosertib (AZD1775) is a potent, small-molecule, ATP-competitive inhibitor of the Wee1 kinase that potentiates the activity of many DNA-damaging chemotherapeutics and is currently in clinical development for multiple indications. The purpose of this study was to investigate the combination of AZD1775 and capecitabine/5FU in preclinical TNBC models. TNBC cell lines were treated with AZD1775 and 5FU and cellular proliferation was assessed in real-time using IncuCyte<sup>&#174;</sup> Live Cell Analysis. Apoptosis was assessed via the Caspase-Glo 3/7 assay system. Western blotting was used to assess changes in expression of downstream effectors. TNBC patient-derived xenograft (PDX) models were treated with AZD1775, capecitabine, or the combination and assessed for tumor growth inhibition. From the initial PDX screen, two of the four TNBC PDX models demonstrated a better response in the combination treatment than either of the single agents. As confirmation, two PDX models were expanded for statistical comparison. Both PDX models demonstrated a significant growth inhibition in the combination versus either of the single agents. (TNBC012, <i>p</i> &lt; 0.05 combo vs. adavosertib or capecitabine, TNBC013, <i>p</i> &lt; 0.01 combo vs. adavosertib or capecitabine.) An enhanced anti-proliferative effect was observed in the adavosertib/5FU combination treatment as measured by live cell analysis. An increase in apoptosis was observed in two of the four cell lines in the combination when compared to single-agent treatment. Treatment with adavosertib as a single agent resulted in a decrease in p-CDC2 in a dose-dependent manner that was also observed in the combination treatment. An increase in &#947;H2AX in two of the four cell lines tested was also observed. No significant changes were observed in Bcl-xL following treatment in any of the cell lines. The combination of adavosertib and capecitabine/5FU demonstrated enhanced combination effects both in vitro and in vivo in preclinical models of TNBC. These results support the clinical investigation of this combination in patients with TNBC, including those with brain metastasis given the CNS penetration of both agents.
first_indexed 2024-03-12T19:59:02Z
format Article
id doaj.art-98303b9316db4c7cb0e1e6133ffcdc79
institution Directory Open Access Journal
issn 2072-6694
language English
last_indexed 2024-03-12T19:59:02Z
publishDate 2020-03-01
publisher MDPI AG
record_format Article
series Cancers
spelling doaj.art-98303b9316db4c7cb0e1e6133ffcdc792023-08-02T02:34:02ZengMDPI AGCancers2072-66942020-03-0112371910.3390/cancers12030719cancers12030719Wee1 Inhibition Enhances the Anti-Tumor Effects of Capecitabine in Preclinical Models of Triple-Negative Breast CancerTodd M. Pitts0Dennis M. Simmons1Stacey M. Bagby2Sarah J. Hartman3Betelehem W. Yacob4Brian Gittleman5John J. Tentler6Diana Cittelly7D. Ryan Ormond8Wells A. Messersmith9S. Gail Eckhardt10Jennifer R. Diamond11Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USADivision of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USADivision of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USADivision of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USADivision of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USADivision of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USADivision of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USADepartment of Pathology, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USADepartment of Neurosurgery, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USADivision of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USADell Medical School, Department of Oncology, The University of Texas Austin, 1701 Trinity Street, Austin, TX 78712, USADivision of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, 12801 E 17th Ave, Aurora, CO 80045, USATriple-negative breast cancer (TNBC) is an aggressive subtype defined by lack of hormone receptor expression and non-amplified HER2. Adavosertib (AZD1775) is a potent, small-molecule, ATP-competitive inhibitor of the Wee1 kinase that potentiates the activity of many DNA-damaging chemotherapeutics and is currently in clinical development for multiple indications. The purpose of this study was to investigate the combination of AZD1775 and capecitabine/5FU in preclinical TNBC models. TNBC cell lines were treated with AZD1775 and 5FU and cellular proliferation was assessed in real-time using IncuCyte<sup>&#174;</sup> Live Cell Analysis. Apoptosis was assessed via the Caspase-Glo 3/7 assay system. Western blotting was used to assess changes in expression of downstream effectors. TNBC patient-derived xenograft (PDX) models were treated with AZD1775, capecitabine, or the combination and assessed for tumor growth inhibition. From the initial PDX screen, two of the four TNBC PDX models demonstrated a better response in the combination treatment than either of the single agents. As confirmation, two PDX models were expanded for statistical comparison. Both PDX models demonstrated a significant growth inhibition in the combination versus either of the single agents. (TNBC012, <i>p</i> &lt; 0.05 combo vs. adavosertib or capecitabine, TNBC013, <i>p</i> &lt; 0.01 combo vs. adavosertib or capecitabine.) An enhanced anti-proliferative effect was observed in the adavosertib/5FU combination treatment as measured by live cell analysis. An increase in apoptosis was observed in two of the four cell lines in the combination when compared to single-agent treatment. Treatment with adavosertib as a single agent resulted in a decrease in p-CDC2 in a dose-dependent manner that was also observed in the combination treatment. An increase in &#947;H2AX in two of the four cell lines tested was also observed. No significant changes were observed in Bcl-xL following treatment in any of the cell lines. The combination of adavosertib and capecitabine/5FU demonstrated enhanced combination effects both in vitro and in vivo in preclinical models of TNBC. These results support the clinical investigation of this combination in patients with TNBC, including those with brain metastasis given the CNS penetration of both agents.https://www.mdpi.com/2072-6694/12/3/719triple-negative breast cancerwee15-fudna damage
spellingShingle Todd M. Pitts
Dennis M. Simmons
Stacey M. Bagby
Sarah J. Hartman
Betelehem W. Yacob
Brian Gittleman
John J. Tentler
Diana Cittelly
D. Ryan Ormond
Wells A. Messersmith
S. Gail Eckhardt
Jennifer R. Diamond
Wee1 Inhibition Enhances the Anti-Tumor Effects of Capecitabine in Preclinical Models of Triple-Negative Breast Cancer
Cancers
triple-negative breast cancer
wee1
5-fu
dna damage
title Wee1 Inhibition Enhances the Anti-Tumor Effects of Capecitabine in Preclinical Models of Triple-Negative Breast Cancer
title_full Wee1 Inhibition Enhances the Anti-Tumor Effects of Capecitabine in Preclinical Models of Triple-Negative Breast Cancer
title_fullStr Wee1 Inhibition Enhances the Anti-Tumor Effects of Capecitabine in Preclinical Models of Triple-Negative Breast Cancer
title_full_unstemmed Wee1 Inhibition Enhances the Anti-Tumor Effects of Capecitabine in Preclinical Models of Triple-Negative Breast Cancer
title_short Wee1 Inhibition Enhances the Anti-Tumor Effects of Capecitabine in Preclinical Models of Triple-Negative Breast Cancer
title_sort wee1 inhibition enhances the anti tumor effects of capecitabine in preclinical models of triple negative breast cancer
topic triple-negative breast cancer
wee1
5-fu
dna damage
url https://www.mdpi.com/2072-6694/12/3/719
work_keys_str_mv AT toddmpitts wee1inhibitionenhancestheantitumoreffectsofcapecitabineinpreclinicalmodelsoftriplenegativebreastcancer
AT dennismsimmons wee1inhibitionenhancestheantitumoreffectsofcapecitabineinpreclinicalmodelsoftriplenegativebreastcancer
AT staceymbagby wee1inhibitionenhancestheantitumoreffectsofcapecitabineinpreclinicalmodelsoftriplenegativebreastcancer
AT sarahjhartman wee1inhibitionenhancestheantitumoreffectsofcapecitabineinpreclinicalmodelsoftriplenegativebreastcancer
AT betelehemwyacob wee1inhibitionenhancestheantitumoreffectsofcapecitabineinpreclinicalmodelsoftriplenegativebreastcancer
AT briangittleman wee1inhibitionenhancestheantitumoreffectsofcapecitabineinpreclinicalmodelsoftriplenegativebreastcancer
AT johnjtentler wee1inhibitionenhancestheantitumoreffectsofcapecitabineinpreclinicalmodelsoftriplenegativebreastcancer
AT dianacittelly wee1inhibitionenhancestheantitumoreffectsofcapecitabineinpreclinicalmodelsoftriplenegativebreastcancer
AT dryanormond wee1inhibitionenhancestheantitumoreffectsofcapecitabineinpreclinicalmodelsoftriplenegativebreastcancer
AT wellsamessersmith wee1inhibitionenhancestheantitumoreffectsofcapecitabineinpreclinicalmodelsoftriplenegativebreastcancer
AT sgaileckhardt wee1inhibitionenhancestheantitumoreffectsofcapecitabineinpreclinicalmodelsoftriplenegativebreastcancer
AT jenniferrdiamond wee1inhibitionenhancestheantitumoreffectsofcapecitabineinpreclinicalmodelsoftriplenegativebreastcancer