1
|
Nakamura A, Tanaka Y, Amano T, Takebayashi A, Takahashi A, Hanada T, Yoneoka Y, Tsuji S, Murakami T. Rapamycin inhibits tamoxifen-induced endometrial proliferation in vitro as a pilot approach for endometrial protection in breast cancer. Sci Rep 2025; 15:2112. [PMID: 39819881 PMCID: PMC11739499 DOI: 10.1038/s41598-025-86586-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/13/2025] [Indexed: 01/19/2025] Open
Abstract
Tamoxifen, a common adjuvant therapy for hormone receptor-positive breast cancer, is associated with an increased risk of endometrial pathologies, such as hyperplasia, polyps, and carcinoma. This study investigates rapamycin, an mTOR inhibitor, as a potential novel strategy for preventing tamoxifen-induced endometrial proliferation. This in vitro study utilised endometrial stromal cells isolated from infertile women. The cells were treated with tamoxifen alone or in combination with rapamycin, and proliferation was assessed using the CCK-8 assay. The activation of the mTOR pathway, as well as apoptosis and cell cycle markers, was evaluated by Western blotting to elucidate the molecular mechanisms underlying these effects. The study design emphasised simulating clinically relevant exposure levels. Tamoxifen significantly increased endometrial cell proliferation in a dose-dependent manner. Rapamycin effectively inhibited this proliferation, even at concentrations lower than those typically observed in clinical settings. Quantitative analysis by Western blotting showed activation of the mTOR pathway and cell cycle in the tamoxifen group, and inhibition of these pathways in the tamoxifen plus rapamycin combination group, whereas there was no change in apoptosis. In conclusion, rapamycin shows promise as a prophylactic agent against tamoxifen-induced endometrial pathologies, with potential implications for fertility preservation and endometrial protection in breast cancer patients.
Collapse
Affiliation(s)
- Akiko Nakamura
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Yuji Tanaka
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan.
| | - Tsukuru Amano
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Akie Takebayashi
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Akimasa Takahashi
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Tetsuro Hanada
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Yutaka Yoneoka
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Shunichiro Tsuji
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Takashi Murakami
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| |
Collapse
|
2
|
Li Z, Chen F, Chen L, Liu J, Tseng D, Hadi F, Omarjee S, Kishore K, Kent J, Kirkpatrick J, D'Santos C, Lawson M, Gertz J, Sikora MJ, McDonnell DP, Carroll JS, Polyak K, Oesterreich S, Lee AV. The EstroGene2.0 database for endocrine therapy response and resistance in breast cancer. NPJ Breast Cancer 2024; 10:106. [PMID: 39702552 PMCID: PMC11659402 DOI: 10.1038/s41523-024-00709-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/08/2024] [Indexed: 12/21/2024] Open
Abstract
Endocrine therapies targeting the estrogen receptor (ER/ESR1) are the cornerstone to treat ER-positive breast cancers patients, but resistance often limits their effectiveness. Notable progress has been made although the fragmented way data is reported has reduced their potential impact. Here, we introduce EstroGene2.0, an expanded database of its precursor 1.0 version. EstroGene2.0 focusses on response and resistance to endocrine therapies in breast cancer models. Incorporating multi-omic profiling of 361 experiments from 212 studies across 28 cell lines, a user-friendly browser offers comprehensive data visualization and metadata mining capabilities ( https://estrogeneii.web.app/ ). Taking advantage of the harmonized data collection, our follow-up meta-analysis revealed transcriptomic landscape and substantial diversity in response to different classes of ER modulators. Endocrine-resistant models exhibit a spectrum of transcriptomic alterations including a contra-directional shift in ER and interferon signalings, which is recapitulated clinically. Dissecting multiple ESR1-mutant cell models revealed the different clinical relevance of cell model engineering and identified high-confidence mutant-ER targets, such as NPY1R. These examples demonstrate how EstroGene2.0 helps investigate breast cancer's response to endocrine therapies and explore resistance mechanisms.
Collapse
Affiliation(s)
- Zheqi Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Fangyuan Chen
- School of Medicine, Tsinghua University, Beijing, China
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Li Chen
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Jiebin Liu
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Danielle Tseng
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Fazal Hadi
- AstraZeneca, The Discovery Centre, Biomedical Campus, Cambridge, UK
| | - Soleilmane Omarjee
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Kamal Kishore
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Joshua Kent
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Joanna Kirkpatrick
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Clive D'Santos
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Mandy Lawson
- AstraZeneca, The Discovery Centre, Biomedical Campus, Cambridge, UK
| | - Jason Gertz
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Matthew J Sikora
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Jason S Carroll
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Steffi Oesterreich
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adrian V Lee
- Women's Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA.
- Institute for Precision Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Serrano D, Johansson H, Bertelsen BE, Gandini S, Mellgren G, Thomas P, Crew KD, Kumar NB, Macis D, Aristarco V, Guerrieri-Gonzaga A, Lazzeroni M, D’Amico M, Buttiron-Webber T, Briata IM, Spinaci S, Galimberti V, Vornik LA, Vilar E, Brown PH, Heckman-Stoddard BM, Szabo E, Bonanni B, DeCensi A. Drug and biomarker tissue levels in a randomized presurgical trial on exemestane alternative schedules. J Natl Cancer Inst 2024; 116:1979-1982. [PMID: 39110531 PMCID: PMC11630545 DOI: 10.1093/jnci/djae183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/19/2024] [Accepted: 07/29/2024] [Indexed: 12/12/2024] Open
Abstract
The drug's activity at the target tissue could help to define the minimal effective dose to promote cancer preventive therapy. Here we present exemestane and sex hormone concentrations within breast tissue from a presurgical study of alternative exemestane schedules. Postmenopausal women candidates for breast surgery for estrogen receptor-positive breast cancer were randomly assigned to exemestane 25 mg once daily (QD), 25 mg 3 times/week (TIW), or 25 mg per week (QW) for 4-6 weeks before surgery. Drug and sex hormones were analyzed from homogenized frozen tissue using a QTRAP 6500+ LC-MS/MS System. Tissue drug concentrations were detectable only in the QD arm with higher concentrations in nonmalignant tissue. Estradiol was nearly suppressed in all groups in the nonmalignant tissue (QD vs TIW P = .364 and QD vs QW P = .693). In contrast, a dose-response trend was observed in cancer tissue. Based on estradiol suppression in nonmalignant tissue, lower exemestane schedules should be explored for breast cancer preventive therapy. Trial Registration: Clinical Trials.gov NCT02598557 and EudraCT 2015-005063-1.
Collapse
Affiliation(s)
- Davide Serrano
- Division of Cancer Prevention and Genetics, European Institute of Oncology IRCCS, Milan, Italy
| | - Harriet Johansson
- Division of Cancer Prevention and Genetics, European Institute of Oncology IRCCS, Milan, Italy
| | - Bjørn-Erik Bertelsen
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Sara Gandini
- Division of Cancer Prevention and Genetics, European Institute of Oncology IRCCS, Milan, Italy
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Parijatham Thomas
- Department of Clinical Cancer Prevention, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Katherine D Crew
- Clinical Breast Cancer Prevention Program, Columbia University Irving Medical Center, New York, NY, USA
| | - Nagi B Kumar
- Departments Cancer Epidemiology, Genitourinary and Breast Oncology, Moffitt Cancer Center, University of South Florida, Tampa, FL, USA
| | - Debora Macis
- Division of Cancer Prevention and Genetics, European Institute of Oncology IRCCS, Milan, Italy
| | - Valentina Aristarco
- Division of Cancer Prevention and Genetics, European Institute of Oncology IRCCS, Milan, Italy
| | | | | | | | | | | | | | - Viviana Galimberti
- Division of Cancer Prevention and Genetics, European Institute of Oncology IRCCS, Milan, Italy
| | - Lana A Vornik
- Department of Clinical Cancer Prevention, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Powel H Brown
- Department of Clinical Cancer Prevention, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Eva Szabo
- Division of Cancer Prevention, NCI, Bethesda, MD, USA
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, European Institute of Oncology IRCCS, Milan, Italy
| | - Andrea DeCensi
- Medical Oncology, Ospedali Galliera, Genoa, Italy
- Wolfson Institute of Population Health, Queen Mary University of London, UK
| |
Collapse
|
4
|
Li Z, Chen F, Chen L, Liu J, Tseng D, Hadi F, Omarjee S, Kishore K, Kent J, Kirkpatrick J, D’Santos C, Lawson M, Gertz J, Sikora MJ, McDonnell DP, Carroll JS, Polyak K, Oesterreich S, Lee AV. EstroGene2.0: A multi-omic database of response to estrogens, ER-modulators, and resistance to endocrine therapies in breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601163. [PMID: 39005294 PMCID: PMC11244912 DOI: 10.1101/2024.06.28.601163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Endocrine therapies targeting the estrogen receptor (ER/ESR1) are the cornerstone to treat ER-positive breast cancers patients, but resistance often limits their effectiveness. Understanding the molecular mechanisms is thus key to optimize the existing drugs and to develop new ER-modulators. Notable progress has been made although the fragmented way data is reported has reduced their potential impact. Here, we introduce EstroGene2.0, an expanded database of its precursor 1.0 version. EstroGene2.0 focusses on response and resistance to endocrine therapies in breast cancer models. Incorporating multi-omic profiling of 361 experiments from 212 studies across 28 cell lines, a user-friendly browser offers comprehensive data visualization and metadata mining capabilities (https://estrogeneii.web.app/). Taking advantage of the harmonized data collection, our follow-up meta-analysis revealed substantial diversity in response to different classes of ER-modulators including SERMs, SERDs, SERCA and LDD/PROTAC. Notably, endocrine resistant models exhibit a spectrum of transcriptomic alterations including a contra-directional shift in ER and interferon signaling, which is recapitulated clinically. Furthermore, dissecting multiple ESR1-mutant cell models revealed the different clinical relevance of genome-edited versus ectopic overexpression model engineering and identified high-confidence mutant-ER targets, such as NPY1R. These examples demonstrate how EstroGene2.0 helps investigate breast cancer's response to endocrine therapies and explore resistance mechanisms.
Collapse
Affiliation(s)
- Zheqi Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Fangyuan Chen
- School of Medicine, Tsinghua University, Beijing, China
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh PA, USA
| | - Li Chen
- Computational Biology Department, Carnegie Mellon University, Pittsburgh PA, USA
| | - Jiebin Liu
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh PA, USA
- Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Danielle Tseng
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh PA, USA
| | | | - Soleilmane Omarjee
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Kamal Kishore
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Joshua Kent
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Joanna Kirkpatrick
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Clive D’Santos
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Jason Gertz
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Matthew J. Sikora
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Donald P. McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Jason S. Carroll
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Steffi Oesterreich
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh PA, USA
| | - Adrian V. Lee
- Women’s Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh PA, USA
- Institute for Precision Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
5
|
Oturkar CC, Rosario SR, Hutson AD, Groman A, Edge SB, Morrison CD, Swetzig WM, Wang J, Park JH, Kaipparettu BA, Singh PK, Kumar S, Cappuccino HH, Ranjan M, Adjei A, Ghasemi M, Goey AK, Kulkarni S, Das GM. ESR1 and p53 interactome alteration defines mechanisms of tamoxifen response in luminal breast cancer. iScience 2024; 27:109995. [PMID: 38868185 PMCID: PMC11166704 DOI: 10.1016/j.isci.2024.109995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 04/25/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024] Open
Abstract
The canonical mechanism behind tamoxifen's therapeutic effect on estrogen receptor α/ESR1+ breast cancers is inhibition of ESR1-dependent estrogen signaling. Although ESR1+ tumors expressing wild-type p53 were reported to be more responsive to tamoxifen (Tam) therapy, p53 has not been factored into choice of this therapy and the mechanism underlying the role of p53 in Tam response remains unclear. In a window-of-opportunity trial on patients with newly diagnosed stage I-III ESR1+/HER2/wild-type p53 breast cancer who were randomized to arms with or without Tam prior to surgery, we reveal that the ESR1-p53 interaction in tumors was inhibited by Tam. This resulted in functional reactivation of p53 leading to transcriptional reprogramming that favors tumor-suppressive signaling, as well as downregulation of oncogenic pathways. These findings illustrating the convergence of ESR1 and p53 signaling during Tam therapy enrich mechanistic understanding of the impact of p53 on the response to Tam therapy.
Collapse
Affiliation(s)
- Chetan C. Oturkar
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Spencer R. Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Alan D. Hutson
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Adrianne Groman
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Stephen B. Edge
- Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Carl D. Morrison
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Wendy M. Swetzig
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jun Hyoung Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | | | - Prashant K. Singh
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Shicha Kumar
- Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Helen H. Cappuccino
- Department of Breast Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Manish Ranjan
- Division of Breast Surgery, Northwestern University Feinberg School of Medicine, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Araba Adjei
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Mohammad Ghasemi
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Andrew K.L. Goey
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Swati Kulkarni
- Division of Breast Surgery, Northwestern University Feinberg School of Medicine, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA
| | - Gokul M. Das
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
6
|
Buijs SM, Koolen SLW, Mathijssen RHJ, Jager A. Tamoxifen Dose De-Escalation: An Effective Strategy for Reducing Adverse Effects? Drugs 2024; 84:385-401. [PMID: 38480629 PMCID: PMC11101371 DOI: 10.1007/s40265-024-02010-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2024] [Indexed: 05/19/2024]
Abstract
Tamoxifen, a cornerstone in the adjuvant treatment of estrogen receptor-positive breast cancer, significantly reduces breast cancer recurrence and breast cancer mortality; however, its standard adjuvant dose of 20 mg daily presents challenges due to a broad spectrum of adverse effects, contributing to high discontinuation rates. Dose reductions of tamoxifen might be an option to reduce treatment-related toxicity, but large randomized controlled trials investigating the tolerability and, more importantly, efficacy of low-dose tamoxifen in the adjuvant setting are lacking. We conducted an extensive literature search to explore evidence on the tolerability and clinical efficacy of reduced doses of tamoxifen. In this review, we discuss two important topics regarding low-dose tamoxifen: (1) the incidence of adverse effects and quality of life among women using low-dose tamoxifen; and (2) the clinical efficacy of low-dose tamoxifen examined in the preventive setting and evaluated through the measurement of several efficacy derivatives. Moreover, practical tools for tamoxifen dose reductions in the adjuvant setting are provided and further research to establish optimal dosing strategies for individual patients are discussed.
Collapse
Affiliation(s)
- Sanne M Buijs
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, PO Box 2040, 3015 CN, Rotterdam, The Netherlands.
| | - Stijn L W Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, PO Box 2040, 3015 CN, Rotterdam, The Netherlands
- Department of Clinical Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, PO Box 2040, 3015 CN, Rotterdam, The Netherlands
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, PO Box 2040, 3015 CN, Rotterdam, The Netherlands
| |
Collapse
|
7
|
Du R, Han X, Deng L, Wang X. Epithelial and mesenchymal phenotypes determine the dynamics of circulating breast tumor cells in microfluidic capillaries under chemotherapy-induced stress. BIOMICROFLUIDICS 2024; 18:024106. [PMID: 38585003 PMCID: PMC10998713 DOI: 10.1063/5.0188861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 03/28/2024] [Indexed: 04/09/2024]
Abstract
Circulating tumor cells (CTCs) with different epithelial and mesenchymal phenotypes play distinct roles in the metastatic cascade. However, the influence of their phenotypic traits and chemotherapy on their transit and retention within capillaries remains unclear. To explore this, we developed a microfluidic device comprising 216 microchannels of different widths from 5 to 16 μm to mimic capillaries. This platform allowed us to study the behaviors of human breast cancer epithelial MCF-7 and mesenchymal MDA-MB-231 cells through microchannels under chemotherapy-induced stress. Our results revealed that when the cell diameter to microchannel width ratio exceeded 1.2, MCF-7 cells exhibited higher transit percentages than MDA-MB-231 cells under a flow rate of 0.13 mm/s. Tamoxifen (250 nM) reduced the transit percentage of MCF-7 cells, whereas 100 nM paclitaxel decreased transit percentages for both cell types. These differential responses were partially due to altered cell stiffness following drug treatments. When cells were entrapped at microchannel entrances, tamoxifen, paclitaxel, and high-flow stress (0.5 mm/s) induced a reduction in mitochondrial membrane potential (MMP) in MCF-7 cells. Tamoxifen treatment also elevated reactive oxygen species (ROS) levels in MCF-7 cells. Conversely, MMP and ROS levels in entrapped MDA-MB-231 cells remained unaffected. Consequently, the viability and proliferation of entrapped MCF-7 cells declined under these chemical and physical stress conditions. Our findings emphasize that phenotypically distinct CTCs may undergo selective filtration and exhibit varied responses to chemotherapy in capillaries, thereby impacting cancer metastasis outcomes. This highlights the importance of considering both cell phenotype and drug response to improve treatment strategies.
Collapse
Affiliation(s)
| | | | - Linhong Deng
- Authors to whom correspondence should be addressed: and
| | - Xiang Wang
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
8
|
Halcrow PW, Quansah DNK, Kumar N, Solloway RL, Teigen KM, Lee KA, Liang B, Geiger JD. Weak base drug-induced endolysosome iron dyshomeostasis controls the generation of reactive oxygen species, mitochondrial depolarization, and cytotoxicity. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2024; 3:33-46. [PMID: 38532786 PMCID: PMC10961484 DOI: 10.1515/nipt-2023-0021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/28/2023] [Indexed: 03/28/2024]
Abstract
Objectives Approximately 75 % of marketed drugs have the physicochemical property of being weak bases. Weak-base drugs with relatively high pKa values enter acidic organelles including endosomes and lysosomes (endolysosomes), reside in and de-acidify endolysosomes, and induce cytotoxicity. Divalent cations within endolysosomes, including iron, are released upon endolysosome de-acidification. Endolysosomes are "master regulators of iron homeostasis", and neurodegeneration is linked to ferrous iron (Fe2+)-induced reactive oxygen species (ROS) generation via Fenton chemistry. Because endolysosome de-acidification-induced lysosome-stress responses release endolysosome Fe2+, it was crucial to determine the mechanisms by which a functionally and structurally diverse group of weak base drugs including atropine, azithromycin, fluoxetine, metoprolol, and tamoxifen influence endolysosomes and cause cell death. Methods Using U87MG astrocytoma and SH-SY5Y neuroblastoma cells, we conducted concentration-response relationships for 5 weak-base drugs to determine EC50 values. From these curves, we chose pharmacologically and therapeutically relevant concentrations to determine if weak-base drugs induced lysosome-stress responses by de-acidifying endolysosomes, releasing endolysosome Fe2+ in sufficient levels to increase cytosolic and mitochondria Fe2+ and ROS levels and cell death. Results Atropine (anticholinergic), azithromycin (antibiotic), fluoxetine (antidepressant), metoprolol (beta-adrenergic), and tamoxifen (anti-estrogen) at pharmacologically and therapeutically relevant concentrations (1) de-acidified endolysosomes, (2) decreased Fe2+ levels in endolysosomes, (3) increased Fe2+ and ROS levels in cytosol and mitochondria, (4) induced mitochondrial membrane potential depolarization, and (5) increased cell death; effects prevented by the endocytosed iron-chelator deferoxamine. Conclusions Weak-base pharmaceuticals induce lysosome-stress responses that may affect their safety profiles; a better understanding of weak-base drugs on Fe2+ interorganellar signaling may improve pharmacotherapeutics.
Collapse
Affiliation(s)
- Peter W. Halcrow
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Darius N. K. Quansah
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Nirmal Kumar
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Rebecca L. Solloway
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Kayla M. Teigen
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Kasumi A. Lee
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Braelyn Liang
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Jonathan D. Geiger
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| |
Collapse
|
9
|
Cai R, Ngwadom C, Saxena R, Soman J, Bruggeman C, Hickey DP, Verduzco R, Ajo-Franklin CM. Creation of a point-of-care therapeutics sensor using protein engineering, electrochemical sensing and electronic integration. Nat Commun 2024; 15:1689. [PMID: 38402222 PMCID: PMC11258353 DOI: 10.1038/s41467-024-45789-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 02/05/2024] [Indexed: 02/26/2024] Open
Abstract
Point-of-care sensors, which are low-cost and user-friendly, play a crucial role in precision medicine by providing quick results for individuals. Here, we transform the conventional glucometer into a 4-hydroxytamoxifen therapeutic biosensor in which 4-hydroxytamoxifen modulates the electrical signal generated by glucose oxidation. To encode the 4-hydroxytamoxifen signal within glucose oxidation, we introduce the ligand-binding domain of estrogen receptor-alpha into pyrroloquinoline quinone-dependent glucose dehydrogenase by constructing and screening a comprehensive protein insertion library. In addition to obtaining 4-hydroxytamoxifen regulatable engineered proteins, these results unveil the significance of both secondary and quaternary protein structures in propagation of conformational signals. By constructing an effective bioelectrochemical interface, we detect 4-hydroxytamoxifen in human blood samples as changes in the electrical signal and use this to develop an electrochemical algorithm to decode the 4-hydroxytamoxifen signal from glucose. To meet the miniaturization and signal amplification requirements for point-of-care use, we harness power from glucose oxidation to create a self-powered sensor. We also amplify the 4-hydroxytamoxifen signal using an organic electrochemical transistor, resulting in milliampere-level signals. Our work demonstrates a broad interdisciplinary approach to create a biosensor that capitalizes on recent innovations in protein engineering, electrochemical sensing, and electrical engineering.
Collapse
Affiliation(s)
- Rong Cai
- Department of Biosciences, Rice University, Houston, TX, USA.
| | | | - Ravindra Saxena
- Applied Physics Graduate Program, Smalley-Curl Institute, Rice University, Houston, TX, USA
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX, USA
| | - Jayashree Soman
- Department of Biosciences, Rice University, Houston, TX, USA
| | - Chase Bruggeman
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, USA
| | - David P Hickey
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, USA
| | - Rafael Verduzco
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX, USA
| | - Caroline M Ajo-Franklin
- Department of Biosciences, Rice University, Houston, TX, USA.
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, TX, USA.
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
10
|
Guishard AR, Guishard AF, Semenova N, Kaushik V, Azad N, Iyer AKV, Yakisich JS. A Short Post-Reattachment Ultrasensitive Window of Time in Human Cancer Cells as Therapeutic Target of Prolonged Low-Dose Administration of Specific Compounds. Int J Cell Biol 2024; 2024:2699572. [PMID: 38352698 PMCID: PMC10861276 DOI: 10.1155/2024/2699572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 11/21/2023] [Accepted: 12/23/2023] [Indexed: 02/16/2024] Open
Abstract
Prolonged low-dose administration (PLDA) of several FDA-approved drugs for noncancer conditions or dietary compounds is associated with a lower incidence of specific types of cancers and with the lower formation of metastasis. However, the underlying mechanism is unknown; there is a discrepancy between the concentration of drugs needed to kill cancer cells in vitro and the actual serum levels (10 and >1000 times lower) found in patients. In this study, we evaluated the hypothesis that clonogenicity may be the target of PLDA. We compared the effect of nigericin (NIG) and menadione (MEN) on the human A549 and H460 lung and MCF-7 and MDA-MB-231 breast cancer cell lines using routine MTT and colony forming assays (CFA). The ability of both NIG and MEN to eliminate 100% of cancer cells was at least 2-10 times more potent in CFA compared to MTT assays. Our results revealed the existence of a short post-reattachment window of time when cancer cells growing at low density are more sensitive to PLDA of specific drugs likely by targeting clonogenic rather than proliferation pathways. This short ultrasensitive window of time (SUSWoT) was cell- and drug-type specific: the SUSWoT for NIG was present in H460, A549, and MDA-MB-231 cells but not evident in MCF-7 cells. Conversely, a similar SUSWoT for MEN was present in MCF-7, MDA-MD-231, and A549 cells but not evident in H460 cells. Our findings partially explain the decreased incidence of specific types of cancer by PLDA of FDA-approved drugs (or dietary compounds) for noncancer conditions.
Collapse
Affiliation(s)
| | | | - Nina Semenova
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton, VA, USA
| | - Vivek Kaushik
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton, VA, USA
| | - Neelam Azad
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton, VA, USA
- Office of the Vice President for Research, Hampton University, Hampton, VA, USA
| | - Anand K. V. Iyer
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton, VA, USA
| | | |
Collapse
|
11
|
Saghafi F, Salehifar E, Ebrahimi P, Shiran MR, Zaboli E, Sohrevardi SM, Jamialahmadi T, Sahebnasagh A, Sahebkar A. Evaluation of the effect of CYP2D6*3, *4,*10, and *17 polymorphisms on the pharmacokinetic of tamoxifen and its metabolites in patients with hormone-positive breast cancer. J Pharm Biomed Anal 2024; 238:115839. [PMID: 37976989 DOI: 10.1016/j.jpba.2023.115839] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/16/2023] [Accepted: 10/31/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND AND OBJECTIVE A high rate of interindividual variability in response to tamoxifen (TAM) in breast cancer patients with CYP2D6 polymorphism has been reported, which affects the patient's therapeutic outcome. The objective of this study was to investigate the pharmacogenomics of CYP2D6 genotyping in Iranian patients with breast cancer treated with adjuvant TAM. METHODS A peripheral blood sample was obtained to determine the steady-state plasma concentrations of TAM and its metabolites (Endoxifen (EN) and 4-Hydroxytamoxifen (4-OHT)) using high-performance liquid chromatography with fluorescence detection (HPLC-FLU) assay. We detected CYP2D6 * 3, * 4, * 10, and * 17 single nucleotide polymorphisms via polymerase chain reaction and restriction fragment length polymorphism (PCR-RFLP) method. RESULTS A total of 84 Iranian estrogen receptor‑positive breast cancer patients receiving the daily dose of 20 mg tamoxifen were recruited. Although a consequent decrease in the median EN and 4-OHT concentrations was observed by comparing poor or intermediate metabolizer patients with an extensive metabolizer population, this difference did not reach a significant level. The mean plasma EN concentrations in poor and intermediate metabolizers were 46.1% (95% CI, 7.4-27.8%) and 59.4% (95% CI, 11.9-37.3%) of extensive metabolizer subjects, respectively. Poor and intermediate metabolizers had the mean plasma 4-OHT concentrations that were 46.6% (95% CI, 0.9-61.7%) and 73.2% (95% CI, 2.7-93.1%) of those of subjects who were extensive metabolizer, respectively. CONCLUSIONS The possible role of genotyping in Iranian patients' response to treatment may explain inter-individual differences in the plasma concentrations of active metabolites of TAM.
Collapse
Affiliation(s)
- Fatemeh Saghafi
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Ebrahim Salehifar
- Department of Clinical Pharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Pouneh Ebrahimi
- Department of Chemistry, Faculty of Sciences, Golestan University, Gorgan, Iran
| | - Mohammad Reza Shiran
- Department of Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ehsan Zaboli
- Department of Internal Medicine, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Mojtaba Sohrevardi
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Adeleh Sahebnasagh
- Clinical Research Center, Department of Internal Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
12
|
Fang Z, Corbizi Fattori G, McKerrell T, Boucher RH, Jackson A, Fletcher RS, Forte D, Martin JE, Fox S, Roberts J, Glover R, Harris E, Bridges HR, Grassi L, Rodriguez-Meira A, Mead AJ, Knapper S, Ewing J, Butt NM, Jain M, Francis S, Clark FJ, Coppell J, McMullin MF, Wadelin F, Narayanan S, Milojkovic D, Drummond MW, Sekhar M, ElDaly H, Hirst J, Paramor M, Baxter EJ, Godfrey AL, Harrison CN, Méndez-Ferrer S. Tamoxifen for the treatment of myeloproliferative neoplasms: A Phase II clinical trial and exploratory analysis. Nat Commun 2023; 14:7725. [PMID: 38001082 PMCID: PMC10673935 DOI: 10.1038/s41467-023-43175-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Current therapies for myeloproliferative neoplasms (MPNs) improve symptoms but have limited effect on tumor size. In preclinical studies, tamoxifen restored normal apoptosis in mutated hematopoietic stem/progenitor cells (HSPCs). TAMARIN Phase-II, multicenter, single-arm clinical trial assessed tamoxifen's safety and activity in patients with stable MPNs, no prior thrombotic events and mutated JAK2V617F, CALRins5 or CALRdel52 peripheral blood allele burden ≥20% (EudraCT 2015-005497-38). 38 patients were recruited over 112w and 32 completed 24w-treatment. The study's A'herns success criteria were met as the primary outcome ( ≥ 50% reduction in mutant allele burden at 24w) was observed in 3/38 patients. Secondary outcomes included ≥25% reduction at 24w (5/38), ≥50% reduction at 12w (0/38), thrombotic events (2/38), toxicities, hematological response, proportion of patients in each IWG-MRT response category and ELN response criteria. As exploratory outcomes, baseline analysis of HSPC transcriptome segregates responders and non-responders, suggesting a predictive signature. In responder HSPCs, longitudinal analysis shows high baseline expression of JAK-STAT signaling and oxidative phosphorylation genes, which are downregulated by tamoxifen. We further demonstrate in preclinical studies that in JAK2V617F+ cells, 4-hydroxytamoxifen inhibits mitochondrial complex-I, activates integrated stress response and decreases pathogenic JAK2-signaling. These results warrant further investigation of tamoxifen in MPN, with careful consideration of thrombotic risk.
Collapse
Affiliation(s)
- Zijian Fang
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Giuditta Corbizi Fattori
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Thomas McKerrell
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Rebecca H Boucher
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Aimee Jackson
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Rachel S Fletcher
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Dorian Forte
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Jose-Ezequiel Martin
- Cancer Molecular Diagnostic Laboratory, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Sonia Fox
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - James Roberts
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Rachel Glover
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Erica Harris
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Hannah R Bridges
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Luigi Grassi
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Alba Rodriguez-Meira
- NIHR Biomedical Research Centre and MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Adam J Mead
- NIHR Biomedical Research Centre and MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | | | - Joanne Ewing
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Nauman M Butt
- The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, UK
| | | | | | - Fiona J Clark
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | | | | | | | | | | | | | - Hesham ElDaly
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Judy Hirst
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Maike Paramor
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
| | - E Joanna Baxter
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Anna L Godfrey
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - Simón Méndez-Ferrer
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
- NHS Blood and Transplant, Cambridge, UK.
| |
Collapse
|
13
|
Sfogliarini C, Pepe G, Cesta CM, Allegretti M, Locati M, Vegeto E. The immune activity of selective estrogen receptor modulators is gene and macrophage subtype-specific yet converges on Il1b downregulation. Biomed Pharmacother 2023; 165:115008. [PMID: 37442065 DOI: 10.1016/j.biopha.2023.115008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/29/2023] [Accepted: 06/11/2023] [Indexed: 07/15/2023] Open
Abstract
Raloxifene belongs to the family of Selective Estrogen Receptor Modulators (SERMs), which are drugs widely prescribed for Estrogen Receptor alpha (ERα)-related pathologies. Recently, SERMs are being tested in repurposing strategies for ERα-independent clinical indications, including a wide range of microbial infections. Macrophages are central in the fight against pathogen invasion. Despite estrogens have been shown to regulate macrophage phenotype, SERMs activity in these cells is still poorly defined. We investigated the activity of Raloxifene in comparison with another widely used SERM, Tamoxifen, on immune gene expression in macrophages obtained from mouse and human tissues, including mouse peritoneal macrophages, bone marrow-derived macrophages, microglia or human blood-derived macrophages, assaying for the involvement of the ERα, PI3K and NRF2 pathways also under inflammatory conditions. Our data demonstrate that Raloxifene acts by a dual mechanism, which entails ERα antagonism and off-target mediators. Moreover, micromolar concentrations of Raloxifene increase the expression of immune metabolic genes, such as Vegfa and Hmox1, through PI3K and NRF2 activation selectively in peritoneal macrophages. Conversely, Il1b mRNA down-regulation by SERMs is consistently observed in all macrophage subtypes and unrelated to the PI3K/NRF2 system. Importantly, the production of the inflammatory cytokine TNFα induced by the bacterial endotoxin, LPS, is potentiated by SERMs and paralleled by the cell subtype-specific increase in IL1β secretion. This work extends our knowledge on the biological and molecular mechanisms of SERMs immune activity and indicate macrophages as a pharmacological target for the exploitation of the antimicrobial potential of these drugs.
Collapse
Affiliation(s)
- Chiara Sfogliarini
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy.
| | - Giovanna Pepe
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy.
| | | | | | - Massimo Locati
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089 Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milan, Italy.
| | - Elisabetta Vegeto
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy.
| |
Collapse
|
14
|
Liu P, Roberts S, Shoemaker JT, Vukasinovic J, Tomlinson DC, Speirs V. Validation of a 3D perfused cell culture platform as a tool for humanised preclinical drug testing in breast cancer using established cell lines and patient-derived tissues. PLoS One 2023; 18:e0283044. [PMID: 36928454 PMCID: PMC10019722 DOI: 10.1371/journal.pone.0283044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 02/28/2023] [Indexed: 03/17/2023] Open
Abstract
3D cell culture models of cancer are currently being developed to recapitulate in vivo physiological conditions and to assess therapeutic responses. However, most models failed to incorporate the biochemical and biophysical stimuli from fluid flow. In this study, a three-dimensional scaffold, SeedEZ was applied within the PerfusionPal perfused culture system to investigate how perfusion, and blood-like oxygen delivery influenced breast cancer cell growth and their responses to a commonly used breast cancer drug tamoxifen. Our results showed that breast cancer cells could be maintained over 3 weeks in PerfusionPal with increased cell viability compared to static 3D culture in fully humanised conditions. This platform also supported examining the effect of tamoxifen on breast cancer cell lines and in primary patient-derived breast cancer samples. Future work is warranted to further the adaption for fully humanised assessment of drug effectiveness in a patient personalized approach with the aim to reduce the burden of animal use in cancer research and increase the degree of human pre-clinical data translation to clinic.
Collapse
Affiliation(s)
- Peng Liu
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Sophie Roberts
- Leeds Institute of Medical Research at St James’s, University of Leeds, Leeds, United Kingdom
| | | | | | - Darren C Tomlinson
- School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Valerie Speirs
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
15
|
Galvano E, Pandit H, Sepulveda J, Ng CAS, Becher MK, Mandelblatt JS, Van Dyk K, Rebeck GW. Behavioral and transcriptomic effects of the cancer treatment tamoxifen in mice. Front Neurosci 2023; 17:1068334. [PMID: 36845433 PMCID: PMC9951777 DOI: 10.3389/fnins.2023.1068334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/24/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction Tamoxifen is a common treatment for estrogen receptor-positive breast cancer. While tamoxifen treatment is generally accepted as safe, there are concerns about adverse effects on cognition. Methods We used a mouse model of chronic tamoxifen exposure to examine the effects of tamoxifen on the brain. Female C57/BL6 mice were exposed to tamoxifen or vehicle control for six weeks; brains of 15 mice were analyzed for tamoxifen levels and transcriptomic changes, and an additional 32 mice were analyzed through a battery of behavioral tests. Results Tamoxifen and its metabolite 4-OH-tamoxifen were found at higher levels in the brain than in the plasma, demonstrating the facile entry of tamoxifen into the CNS. Behaviorally, tamoxifen-exposed mice showed no impairment in assays related to general health, exploration, motor function, sensorimotor gating, and spatial learning. Tamoxifen-treated mice showed a significantly increased freezing response in a fear conditioning paradigm, but no effects on anxiety measures in the absence of stressors. RNA sequencing analysis of whole hippocampi showed tamoxifen-induced reductions in gene pathways related to microtubule function, synapse regulation, and neurogenesis. Discussion These findings of the effects of tamoxifen exposure on fear conditioning and on gene expression related to neuronal connectivity suggest that there may be CNS side effects of this common breast cancer treatment.
Collapse
Affiliation(s)
- Elena Galvano
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Harshul Pandit
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Jordy Sepulveda
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Christi Anne S. Ng
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Melanie K. Becher
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| | - Jeanne S. Mandelblatt
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, United States
| | - Kathleen Van Dyk
- Department of Psychiatry, UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, United States
| | - G. William Rebeck
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
16
|
Ayeni OA, Chiwambutsa S, Chen WC, Kapungu N, Kanji C, Thelingwani R, Murugan N, Mathiba R, Phakathi B, Nietz S, Ramiah D, O'Neil DS, Jacobson JS, Ruff P, Cubasch H, Chirwa T, Joffe M, Masimirembwa C, Neugut AI. The impact of HIV on non-adherence for tamoxifen among women with breast cancer in South Africa. Breast Cancer Res Treat 2023; 197:647-659. [PMID: 36538247 PMCID: PMC10149344 DOI: 10.1007/s10549-022-06835-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE Women living with HIV (WLWH) and breast cancer (BC) have worse overall survival than HIV-negative women with BC, and poor adherence to prescribed tamoxifen is known to contribute to poor survival. We therefore investigated the association of HIV infection with adherence to adjuvant tamoxifen among women with localized hormone receptor (HR)-positive breast cancer in South Africa. METHODS Among 4,097 women diagnosed with breast cancer at six hospitals in the prospective South African Breast Cancer and HIV Outcomes (SABCHO) cohort study between July 2015 and December 2020, we focused on black women with stages I-III HR-positive breast cancer who were prescribed 20 mg of adjuvant tamoxifen daily. We collected venous blood once from each participant during a routine clinic visit, and analyzed concentrations of tamoxifen and its metabolites using a triple quadruple mass spectrometer. We defined non-adherence as a tamoxifen level < 60 ng/mL after 3 months of daily tamoxifen use. We compared tamoxifen-related side effects, and concurrent medication use among women with and without HIV and developed multivariable logistic regression models of tamoxifen non-adherence. RESULTS Among 369 subjects, 78 (21.1%) were WLWH and 291 (78.9%) were HIV-negative. After a median (interquartile range) time of 13.0 (6.2-25.2) months since tamoxifen initiation, the tamoxifen serum concentration ranged between 1.54 and 943.0 ng/mL and 208 (56.4%) women were non-adherent to tamoxifen. Women < 40 years of age were more likely to be non-adherent than women > 60 years (73.4% vs 52.6%, odds ratio (OR) = 2.49, 95% confidence interval (CI) = 1.26-4.94); likewise, WLWH (70.5% vs 52.6%, OR = 2.16, 95% CI = 1.26-3.70) than HIV-negative women. In an adjusted model WLWH had twice the odds of non-adherence to tamoxifen, compared to HIV-negative women (OR = 2.40, 95% CI = 1.11-5.20). CONCLUSION High rates of non-adherence to adjuvant tamoxifen may limit the overall survival of black South African women with HR-positive breast cancer, especially among WLWH.
Collapse
Affiliation(s)
- Oluwatosin A Ayeni
- MRC South Africa and the University of the Witwatersrand Centre for Common Epithelial Cancers Research Unit, Johannesburg, South Africa.
- Soweto Comprehensive Cancer Centre, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa.
| | - Shingirai Chiwambutsa
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Wenlong Carl Chen
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
| | - Nyasha Kapungu
- Department of Pharmaceutical Medicine, African Institute of Biomedical Science and Technology Block C Wilkins Hospital, Harare, Zimbabwe
| | - Comfort Kanji
- Department of Pharmaceutical Medicine, African Institute of Biomedical Science and Technology Block C Wilkins Hospital, Harare, Zimbabwe
| | - Roslyn Thelingwani
- Department of Pharmaceutical Medicine, African Institute of Biomedical Science and Technology Block C Wilkins Hospital, Harare, Zimbabwe
| | - Nivashni Murugan
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Surgery, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Rofhiwa Mathiba
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Soweto Comprehensive Cancer Centre, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - Boitumelo Phakathi
- Department of Surgery, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sarah Nietz
- Department of Surgery, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Duvern Ramiah
- Department of Radiation Oncology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Daniel S O'Neil
- Sylvester Comprehensive Cancer Center and Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Judith S Jacobson
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Paul Ruff
- MRC South Africa and the University of the Witwatersrand Centre for Common Epithelial Cancers Research Unit, Johannesburg, South Africa
- Soweto Comprehensive Cancer Centre, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
| | - Herbert Cubasch
- Department of Surgery, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Tobias Chirwa
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, 27 St Andrews Road, Parktown, Johannesburg, 2193, South Africa
| | - Maureen Joffe
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- MRC South Africa and the University of the Witwatersrand Centre for Common Epithelial Cancers Research Unit, Johannesburg, South Africa
| | - Collen Masimirembwa
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Pharmaceutical Medicine, African Institute of Biomedical Science and Technology Block C Wilkins Hospital, Harare, Zimbabwe
| | - Alfred I Neugut
- Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| |
Collapse
|
17
|
Hagan ML, Mander S, Joseph C, Mcgrath M, Barrett A, Lewis A, Hill WD, Browning D, Mcgee-Lawrence ME, Cai H, Liu K, Barrett JT, Gewirtz DA, Thangaraju M, Schoenlein PV. Upregulation of the EGFR/MEK1/MAPK1/2 signaling axis as a mechanism of resistance to antiestrogen‑induced BimEL dependent apoptosis in ER + breast cancer cells. Int J Oncol 2022; 62:20. [PMID: 36524361 PMCID: PMC9854236 DOI: 10.3892/ijo.2022.5468] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is commonly upregulated in multiple cancer types, including breast cancer. In the present study, evidence is provided in support of the premise that upregulation of the EGFR/MEK1/MAPK1/2 signaling axis during antiestrogen treatment facilitates the escape of breast cancer cells from BimEL‑dependent apoptosis, conferring resistance to therapy. This conclusion is based on the findings that ectopic BimEL cDNA overexpression and confocal imaging studies confirm the pro‑apoptotic role of BimEL in ERα expressing breast cancer cells and that upregulated EGFR/MEK1/MAPK1/2 signaling blocks BimEL pro‑apoptotic action in an antiestrogen‑resistant breast cancer cell model. In addition, the present study identified a pro‑survival role for autophagy in antiestrogen resistance while EGFR inhibitor studies demonstrated that a significant percentage of antiestrogen‑resistant breast cancer cells survive EGFR targeting by pro‑survival autophagy. These pre‑clinical studies establish the possibility that targeting both the MEK1/MAPK1/2 signaling axis and pro‑survival autophagy may be required to eradicate breast cancer cell survival and prevent the development of antiestrogen resistance following hormone treatments. The present study uniquely identified EGFR upregulation as one of the mechanisms breast cancer cells utilize to evade the cytotoxic effects of antiestrogens mediated through BimEL‑dependent apoptosis.
Collapse
Affiliation(s)
- Mackenzie L. Hagan
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA 30912, USA
| | - Suchreet Mander
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA 30912, USA
| | - Carol Joseph
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA 30912, USA
| | - Michael Mcgrath
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA 30912, USA
| | - Amanda Barrett
- Department of Pathology, Augusta University, Augusta, GA 30912, USA,Department of Medical College of Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Allison Lewis
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA 30912, USA
| | - William D. Hill
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Darren Browning
- Department of Medical College of Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA,Department of Biochemistry, Augusta University, Augusta, GA 30912, USA
| | | | - Haifeng Cai
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA 30912, USA,Department of Surgical Oncology, Tangshan People's Hospital, Tangshan, Hebei 063000, P.R. China
| | - Kebin Liu
- Department of Medical College of Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA,Department of Biochemistry, Augusta University, Augusta, GA 30912, USA
| | - John T. Barrett
- Department of Medical College of Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA,Department of Radiation Oncology, Augusta University, Augusta, GA 30912, USA
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, Massey Cancer Center, Richmond, VA 23298, USA
| | - Muthusamy Thangaraju
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA 30912, USA,Department of Medical College of Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA
| | - Patricia V. Schoenlein
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA 30912, USA,Department of Medical College of Georgia Cancer Center, Augusta University, Augusta, GA 30912, USA,Correspondence to: Dr Patricia V. Schoenlein, Department of Cellular Biology and Anatomy, Augusta University, Research and Education Building Room 2912, 1120 15th Street, Augusta, GA 30912, USA, E-mail:
| |
Collapse
|
18
|
Francis JWM, Saundh M, Parks RM, Cheung KL. Molecular Biomarker Expression in Window of Opportunity Studies for Oestrogen Receptor Positive Breast Cancer-A Systematic Review of the Literature. Cancers (Basel) 2022; 14:cancers14205027. [PMID: 36291809 PMCID: PMC9599781 DOI: 10.3390/cancers14205027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Window of opportunity (WoO) trials allow the opportunity to assess the use of drugs in breast cancer research before treatment has commenced. The aim of this review of the literature is to review WoO trials in patients with oestrogen receptor-positive (ER+) breast cancer to help guide treatment. This will be useful for patients who receive drug treatment before surgery, or as an alternative to surgery in older, more frail adults. Abstract Window of opportunity (WoO) trials create the opportunity to demonstrate pharmacodynamic parameters of a drug in vivo and have increasing use in breast cancer research. Most breast cancer tumours are oestrogen receptor-positive (ER+), leading to the development of multiple treatment options tailored towards this particular tumour subtype. The aim of this literature review is to review WoO trials pertaining to the pharmacodynamic activity of drugs available for use in ER+ breast cancer in order to help guide treatment for patients receiving neoadjuvant and primary endocrine therapy. Five databases (EMBASE, Cochrane, MEDLINE, PubMed, Web of Science) were searched for eligible studies. Studies performed in treatment-naïve patients with histologically confirmed ER+ breast cancer were included if they acquired pre- and post-treatment biopsies, compared measurement of a proteomic biomarker between these two biopsies and delivered treatment for a maximum mean duration of 31 days. Fifteen studies were eligible for inclusion and covered six different drug classes: three endocrine therapies (ETs) including aromatase inhibitors (AIs), selective oestrogen receptor modulators (SERMs), selective oestrogen receptor degraders (SERDs) and three non-ETs including mTOR inhibitors, AKT inhibitors and synthetic oestrogens. Ki67 was the most frequently measured marker, appearing in all studies. Progesterone receptor (PR) and ER were the next most frequently measured markers, appearing five and four studies, respectively. All three of these markers were significantly downregulated in both AIs and SERDs; Ki67 alone was downregulated in SERMs. Less commonly assessed markers including pS6, pGSH3B, FSH and IGF1 were downregulated while CD34, pAKT and SHBG were significantly upregulated. There were no significant changes in the other biomarkers measured such as phosphate and tensin homolog (PTEN), Bax and Bcl-2.WoO studies have been widely utilised within the ER+ breast cancer subtype, demonstrating their worth in pharmacodynamic research. However, research remains focused upon routinely measured biomarkers such ER PR and Ki67, with an array of less common markers sporadically used.
Collapse
Affiliation(s)
- James W. M. Francis
- School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- Nottingham Breast Cancer Research Centre, University of Nottingham, Nottingham NG7 2RD, UK
| | - Manmeet Saundh
- School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- Nottingham Breast Cancer Research Centre, University of Nottingham, Nottingham NG7 2RD, UK
| | - Ruth M. Parks
- School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- Nottingham Breast Cancer Research Centre, University of Nottingham, Nottingham NG7 2RD, UK
| | - Kwok-Leung Cheung
- School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- Nottingham Breast Cancer Research Centre, University of Nottingham, Nottingham NG7 2RD, UK
- Correspondence:
| |
Collapse
|
19
|
Bacterial diet modulates tamoxifen-induced death via host fatty acid metabolism. Nat Commun 2022; 13:5595. [PMID: 36151093 PMCID: PMC9508336 DOI: 10.1038/s41467-022-33299-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 09/06/2022] [Indexed: 11/18/2022] Open
Abstract
Tamoxifen is a selective estrogen receptor (ER) modulator that is used to treat ER-positive breast cancer, but that at high doses kills both ER-positive and ER-negative breast cancer cells. We recapitulate this off-target effect in Caenorhabditis elegans, which does not have an ER ortholog. We find that different bacteria dramatically modulate tamoxifen toxicity in C. elegans, with a three-order of magnitude difference between animals fed Escherichia coli, Comamonas aquatica, and Bacillus subtilis. Remarkably, host fatty acid (FA) biosynthesis mitigates tamoxifen toxicity, and different bacteria provide the animal with different FAs, resulting in distinct FA profiles. Surprisingly these bacteria modulate tamoxifen toxicity by different death mechanisms, some of which are modulated by FA supplementation and others by antioxidants. Together, this work reveals a complex interplay between microbiota, FA metabolism and tamoxifen toxicity that may provide a blueprint for similar studies in more complex mammals. Here, Diot et al. use the nematode Caenorhabditis elegans as a model to identify off-target toxicity mechanisms for tamoxifen, and find that these include fatty acid metabolism and cell death, which can be modulated by different bacterial species.
Collapse
|
20
|
Evaluation of human adipose-derived stromal cell behaviour following exposure to Tamoxifen. Tissue Cell 2022; 77:101858. [DOI: 10.1016/j.tice.2022.101858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 11/18/2022]
|
21
|
Calhoun S, Duan L, Maki CG. Acetyl-CoA synthetases ACSS1 and ACSS2 are 4-hydroxytamoxifen responsive factors that promote survival in tamoxifen treated and estrogen deprived cells. Transl Oncol 2022; 19:101386. [PMID: 35263700 PMCID: PMC8904238 DOI: 10.1016/j.tranon.2022.101386] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 12/22/2022] Open
Affiliation(s)
- Sarah Calhoun
- Department of Anatomy and Cell Biology, Rush University Medical Center, 600 S. Paulina Ave, AcFac Suite 507, Chicago, IL 60612, USA
| | - Lei Duan
- Department of Anatomy and Cell Biology, Rush University Medical Center, 600 S. Paulina Ave, AcFac Suite 507, Chicago, IL 60612, USA
| | - Carl G Maki
- Department of Anatomy and Cell Biology, Rush University Medical Center, 600 S. Paulina Ave, AcFac Suite 507, Chicago, IL 60612, USA.
| |
Collapse
|
22
|
Limniatis G, Georges E. Down-regulation of ABCB1 by Collateral Sensitivity Drugs Reverses Multidrug Resistance and Up-regulates Enolase I. J Biochem 2022; 172:37-48. [PMID: 35471238 DOI: 10.1093/jb/mvac032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 04/04/2022] [Indexed: 11/12/2022] Open
Abstract
The emergence of drug resistance remains an obstacle in the clinical treatment of cancer. Recent developments in the studies of drug resistance have identified compounds such as verapamil and tamoxifen that specifically target ABCB1-expressing multidrug resistant (MDR) cells, through an ATP-dependent ROS-generating mechanism. In this report, we demonstrate that treatment of ABCB1-expressing multidrug resistant cells (CHORC5 or MDA-Doxo400) or individual clones of the latter with sub-lethal concentrations of tamoxifen or verapamil down-regulates ABCB1 protein and mRNA expression in surviving clones. Consequently, tamoxifen- and verapamil-treated cells show increased sensitivity to chemotherapeutic drugs (e.g., colchicine and doxorubicin) and decreased sensitivity to collateral sensitivity drugs (e.g., verapamil and tamoxifen). Importantly, we show for the first time that down-regulation of ABCB1 expression resulting from tamoxifen treatment and CRISPR-knockout of ABCB1 expression up-regulate α-enolase (enolase I) protein levels and activity. These findings demonstrate a possible effect of ABCB1 expression on the metabolic homeostasis of MDR cells. Moreover, given the use of tamoxifen to prevent the recurrence of estrogen receptor-positive breast cancer, the findings of this study may be clinically important in modulating activity of other drugs.
Collapse
Affiliation(s)
| | - Elias Georges
- Institute of Parasitology, McGill University, Quebec, Canada
| |
Collapse
|
23
|
Sfogliarini C, Pepe G, Dolce A, Della Torre S, Cesta MC, Allegretti M, Locati M, Vegeto E. Tamoxifen Twists Again: On and Off-Targets in Macrophages and Infections. Front Pharmacol 2022; 13:879020. [PMID: 35431927 PMCID: PMC9006819 DOI: 10.3389/fphar.2022.879020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/08/2022] [Indexed: 11/25/2022] Open
Abstract
Beyond the wide use of tamoxifen in breast cancer chemotherapy due to its estrogen receptor antagonist activity, this drug is being assayed in repurposing strategies against a number of microbial infections. We conducted a literature search on the evidence related with tamoxifen activity in macrophages, since these immune cells participate as a first line-defense against pathogen invasion. Consistent data indicate the existence of estrogen receptor-independent targets of tamoxifen in macrophages that include lipid mediators and signaling pathways, such as NRF2 and caspase-1, which allow these cells to undergo phenotypic adaptation and potentiate the inflammatory response, without the induction of cell death. Thus, these lines of evidence suggest that the widespread antimicrobial activity of this drug can be ascribed, at least in part, to the potentiation of the host innate immunity. This widens our understanding of the pharmacological activity of tamoxifen with relevant therapeutic implications for infections and other clinical indications that may benefit from the immunomodulatory effects of this drug.
Collapse
Affiliation(s)
- Chiara Sfogliarini
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Giovanna Pepe
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Arianna Dolce
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Sara Della Torre
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | | | | | - Massimo Locati
- IRCCS Humanitas Research Hospital, Rozzano, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Elisabetta Vegeto
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
24
|
Due SL, Watson DI, Bastian I, Eichelmann AK, Hussey DJ. Oestrogen Receptor Isoforms May Represent a Therapeutic Target in Oesophageal Adenocarcinoma. Cancers (Basel) 2022; 14:1891. [PMID: 35454796 PMCID: PMC9032750 DOI: 10.3390/cancers14081891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/18/2022] [Accepted: 03/26/2022] [Indexed: 02/01/2023] Open
Abstract
Oesophageal adenocarcinoma is a rapidly increasing problem in which treatment options are limited. Previous studies have shown that oesophageal adenocarcinoma cells and tissues express oestrogen receptors (ERs) and show growth suppression and apoptosis in response to ER modulator agents such as tamoxifen. ERs are known to be expressed in a number of isoforms that act together to regulate cell growth and cell death. In this study, we used western blotting to profile the expression of ERα and ERβ isoforms, and expression of the oncologically related molecules p53, HER2, and EGFR, in a panel of oesophageal adenocarcinoma cell lines. The cytotoxicity of tamoxifen in the cell lines was determined with Annexin V-FITC flow cytometry, and correlations between cytotoxicity and receptor expression were assessed using Spearman's rank-order correlation. Oesophageal adenocarcinoma cell lines showed varying cytotoxicity in response to tamoxifen. The ER species ERα90, ERα50, and ERα46, as well as p53, were positively associated with a cytotoxic response. Conversely, ERα74, ERα70, and ERβ54 were associated with a lack of cytotoxic response. The ER species detected in oesophageal adenocarcinoma cells may work together to confer sensitivity to ER modulators in this disease, which could open up a new avenue for therapy in selected patients.
Collapse
Affiliation(s)
- Steven L. Due
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA 5042, Australia; (S.L.D.); (D.I.W.)
- Flinders Health and Medical Research Institute—Cancer Program, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (I.B.); (A.-K.E.)
| | - David I. Watson
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA 5042, Australia; (S.L.D.); (D.I.W.)
- Flinders Health and Medical Research Institute—Cancer Program, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (I.B.); (A.-K.E.)
| | - Isabell Bastian
- Flinders Health and Medical Research Institute—Cancer Program, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (I.B.); (A.-K.E.)
| | - Ann-Kathrin Eichelmann
- Flinders Health and Medical Research Institute—Cancer Program, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (I.B.); (A.-K.E.)
| | - Damian J. Hussey
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA 5042, Australia; (S.L.D.); (D.I.W.)
- Flinders Health and Medical Research Institute—Cancer Program, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia; (I.B.); (A.-K.E.)
| |
Collapse
|
25
|
Razmovski-Naumovski V, Kimble B, Laurenti D, Nammi S, Norimoto H, Chan K. Polysaccharide Peptide Extract From Coriolus versicolor Increased Tmax of Tamoxifen and Maintained Biochemical Serum Parameters, With No Change in the Metabolism of Tamoxifen in the Rat. Front Pharmacol 2022; 13:857864. [PMID: 35450034 PMCID: PMC9016780 DOI: 10.3389/fphar.2022.857864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/07/2022] [Indexed: 11/22/2022] Open
Abstract
Background: Polysaccharide peptide (PSP) extract of Coriolus versicolor (L.) Quél. (1886) (Trametes; Polyporaceae) is increasingly used in cancer to support the immune system. However, its interaction with tamoxifen is unknown. Aim of the study: To investigate the effect of a PSP extract on the pharmacokinetics, biochemical parameters, and depletion of tamoxifen. Methods: The pharmacokinetic and biochemical parameters of tamoxifen (20 mg/mL oral single dose and repeated dosing for 12 days) was investigated in female Sprague Dawley rats with or without PSP (340 mg/kg orally for 7 days) (n = 5 per group). Tamoxifen (5 µM) depletion rate with PSP (10–100 μg/mL) was measured in female rat hepatic microsomes in vitro. Results: Compared to tamoxifen alone, the time to reach maximum concentration (Tmax) significantly increased by 228% (4.15 ± 1.15 versus 13.6 ± 2.71 h) in the single tamoxifen dose with PSP and 93% (6 ± 2.17 versus 11.6 ± 0.4 h) in the repeated tamoxifen dosing with PSP (p < 0.05). No significant changes in the area-under-curve and maximum concentration were observed in the single dose and repeated tamoxifen dosing plus PSP compared to tamoxifen alone. Pharmacodynamically, the repeated tamoxifen dosing with PSP maintained 19 out of 23 hepatic, renal and cardiac biochemical serum parameters in rats compared to untreated rats (p > 0.05). PSP extract did not significantly alter in vitro intrinsic clearance of tamoxifen compared to tamoxifen control. Conclusion: With the increased use of PSP as an adjunct therapy, this study highlights the importance of clinician’s knowledge of its interaction with tamoxifen to avoid compromising clinical actions and enhancing clinical therapy.
Collapse
Affiliation(s)
- Valentina Razmovski-Naumovski
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
- School of Science, Western Sydney University, Penrith, NSW, Australia
- South West Sydney Clinical Campuses, Discipline of Medicine, University of New South Wales Sydney, Sydney, NSW, Australia
- *Correspondence: Valentina Razmovski-Naumovski, ; Srinivas Nammi, ; Kelvin Chan,
| | - Benjamin Kimble
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
| | - Daunia Laurenti
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
- School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Srinivas Nammi
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
- School of Science, Western Sydney University, Penrith, NSW, Australia
- *Correspondence: Valentina Razmovski-Naumovski, ; Srinivas Nammi, ; Kelvin Chan,
| | - Hisayoshi Norimoto
- R&D Centre of PuraPharm Corporation Ltd. and PuraPharm (Nanning) Pharmaceutical Co. Ltd., Hong Kong, Hong Kong SAR, China
| | - Kelvin Chan
- NICM Health Research Institute, Western Sydney University, Penrith, NSW, Australia
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
- *Correspondence: Valentina Razmovski-Naumovski, ; Srinivas Nammi, ; Kelvin Chan,
| |
Collapse
|
26
|
Unten Y, Murai M, Koshitaka T, Kitao K, Shirai O, Masuya T, Miyoshi H. Comprehensive understanding of multiple actions of anticancer drug tamoxifen in isolated mitochondria. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2022; 1863:148520. [PMID: 34896079 DOI: 10.1016/j.bbabio.2021.148520] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/11/2021] [Accepted: 12/02/2021] [Indexed: 12/13/2022]
Abstract
Tamoxifen has been widely used in the treatment of estrogen receptor (ER)-positive breast cancer, whereas it also exhibits ER-independent anticancer effects in various cancer cell types. As one of the convincing mechanisms underlying the ER-independent effects, induction of apoptosis through mitochondrial dysfunction has been advocated. However, the mechanism of action of tamoxifen even at the isolated mitochondrial level is not fully understood and remains controversial. Here, we attempted to comprehensively understand tamoxifen's multiple actions in isolated rat liver mitochondria through not only revisiting the actions hitherto reported but also conducting originally designed experiments. Using submitochondrial particles, we found that tamoxifen has potential as an inhibitor of both respiratory complex I and ATP synthase. However, these inhibitory effects were not elicited in intact mitochondria, likely because penetration of tamoxifen across the inner mitochondrial membrane is highly restricted owing to its localized positive charge (-N+H(CH3)2). This restricted penetration may also explain why tamoxifen is unable to function as a protonophore-type uncoupler in mitochondria. Moreover, tamoxifen suppressed opening of the mitochondrial permeability transition pore induced by Ca2+ overload through enhancing phosphate uptake into the matrix. The photoaffinity labeling experiments using a photolabile tamoxifen derivative (pTAM1) indicated that pTAM1 specifically binds to voltage-dependent anion channels (VDACs) 1 and 3, which regulate transport of various substances into mitochondria. The binding of tamoxifen to VDAC1 and/or VDAC3 could be responsible for the enhancement of phosphate uptake. Taking all the results together, we consider the principal impairment of mitochondrial functions caused by tamoxifen.
Collapse
Affiliation(s)
- Yufu Unten
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Masatoshi Murai
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Tomoki Koshitaka
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Kotaro Kitao
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Osamu Shirai
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Takahiro Masuya
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Hideto Miyoshi
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| |
Collapse
|
27
|
Miranda C, Galleguillos M, Torres R, Tardón K, Cáceres DD, Lee K, Redal MA, Varela NM, Quiñones LA. Preliminary Pharmacogenomic-Based Predictive Models of Tamoxifen Response in Hormone-dependent Chilean Breast Cancer Patients. Front Pharmacol 2021; 12:661443. [PMID: 34899282 PMCID: PMC8656167 DOI: 10.3389/fphar.2021.661443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022] Open
Abstract
Tamoxifen (TAM), a selective oestrogen receptor modulator, is one of the most used treatments in oestrogen receptor-positive (ER+) early and metastatic breast cancer (BC) patients. The response to TAM has a high degree of inter-individual variability. This is mainly due to genetic variants in CYP2D6 gene, as well as other genes encoding proteins involved in the TAM pharmacokinetic and/or pharmacodynamic. Therefore, prediction of the TAM response using these genetic factors together with other non-genetic variables may be relevant to improve breast cancer treatment. Thus, in this work, we used genetic polymorphisms and clinical variables for TAM response modelling. One hundred sixty-two ER + BC patients with 2 years of TAM treatment were retrospectively recruited, and the genetic polymorphisms CYP2D6*4, CYP3A4*1B (CYP3A4*1.001), CYP3A5*3, UGT2B7*2, UGT2B15*2, SULT1A1*2, and ESRA V364E were analyzed by PCR-RFLP. Concomitantly, the therapeutic response was obtained from clinical records for association with genotypes using univariate and multivariate biostatistical models. Our results show that UGT2B15*1/*2 genotype protects against relapse (OR = 0.09; p = 0.02), CYP3A5*3/*3 genotype avoids endometrial hyperplasia (OR = 0.07; p = 0.01), SULT1A1*1/*2 genotype avoids vaginal bleeding (OR = 0.09; p = 0.03) and ESRA 364E/364E genotype increases the probability of vaginal bleeding (OR = 5.68; p = 0.02). Logistic regression models, including genomic and non-genomic variables, allowed us to obtain preliminary predictive models to explain relapse (p = 0.010), endometrial hyperplasia (p = 0.002) and vaginal bleeding (p = 0.014). Our results suggest that the response to TAM treatment in ER + BC patients might be associated with the presence of the studied genetic variants in UGT2B15, CYP3A5, SULT1A1 and ESRA genes. After clinical validation protocols, these models might be used to help to predict a percentage of BC relapse and adverse reactions, improving the individual response to TAM-based treatment.
Collapse
Affiliation(s)
- Carla Miranda
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Macarena Galleguillos
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago, Chile
| | | | | | - Dante D Cáceres
- Institute of Population Health, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Kuen Lee
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago, Chile.,Faculty of Medicine, University of Chile, Santiago, Chile
| | - María A Redal
- Genetic Division, Department of Medicine, Hospital de Clínicas José de San Martín, Buenos Aires, Argentina.,Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | - Nelson M Varela
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago, Chile.,Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| | - Luis A Quiñones
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago, Chile.,Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain
| |
Collapse
|
28
|
Pepe G, Sfogliarini C, Rizzello L, Battaglia G, Pinna C, Rovati G, Ciana P, Brunialti E, Mornata F, Maggi A, Locati M, Vegeto E. ERα-independent NRF2-mediated immunoregulatory activity of tamoxifen. Biomed Pharmacother 2021; 144:112274. [PMID: 34653752 DOI: 10.1016/j.biopha.2021.112274] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/30/2022] Open
Abstract
Sex differences in immune-mediated diseases are linked to the activity of estrogens on innate immunity cells, including macrophages. Tamoxifen (TAM) is a selective estrogen receptor modulator (SERM) used in estrogen receptor-alpha (ERα)-dependent breast cancers and off-target indications such as infections, although the immune activity of TAM and its active metabolite, 4-OH tamoxifen (4HT), is poorly characterized. Here, we aimed at investigating the endocrine and immune activity of these SERMs in macrophages. Using primary cultures of female mouse macrophages, we analyzed the expression of immune mediators and activation of effector functions in competition experiments with SERMs and 17β-estradiol (E2) or the bacterial endotoxin LPS. We observed that 4HT and TAM induce estrogen antagonist effects when used at nanomolar concentrations, while pharmacological concentrations that are reached by TAM in clinical settings regulate the expression of VEGFα and other immune activation genes by ERα- and G protein-coupled receptor 1 (GPER1)-independent mechanisms that involve NRF2 through PI3K/Akt-dependent mechanisms. Importantly, we observed that SERMs potentiate cell phagocytosis and modify the effects of LPS on the expression of inflammatory cytokines, such as TNFα and IL1β, with an overall increase in cell inflammatory phenotype, further sustained by potentiation of IL1β secretion through caspase-1 activation. Altogether, our data unravel a novel molecular mechanism and immune functions for TAM and 4HT, sustaining their repurposing in infective and other estrogen receptors-unrelated pathologies.
Collapse
Affiliation(s)
- Giovanna Pepe
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Chiara Sfogliarini
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Loris Rizzello
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy; National Institute of Molecular Genetics (INGM) Milan, 20122 Milan, Italy
| | - Giuseppe Battaglia
- Department of Chemistry and; The EPSRC/Jeol Centre for Liquid Phase Electron Microscopy, University College London, WC1H 0AJ London, U.K; Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain; Institute for Physics of Living System, University College London, WC1E 6BT London, U.K; Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| | - Christian Pinna
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Gianenrico Rovati
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Paolo Ciana
- Department of Health Sciences University of Milan, 20142 Milan, Italy
| | - Electra Brunialti
- Department of Health Sciences University of Milan, 20142 Milan, Italy
| | - Federica Mornata
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Adriana Maggi
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy
| | - Massimo Locati
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20133 Milan, Italy
| | - Elisabetta Vegeto
- Department of Pharmaceutical Sciences, University of Milan, 20133 Milan, Italy.
| |
Collapse
|
29
|
Petri BJ, Piell KM, South Whitt GC, Wilt AE, Poulton CC, Lehman NL, Clem BF, Nystoriak MA, Wysoczynski M, Klinge CM. HNRNPA2B1 regulates tamoxifen- and fulvestrant-sensitivity and hallmarks of endocrine resistance in breast cancer cells. Cancer Lett 2021; 518:152-168. [PMID: 34273466 PMCID: PMC8358706 DOI: 10.1016/j.canlet.2021.07.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 12/31/2022]
Abstract
Despite new combination therapies improving survival of breast cancer patients with estrogen receptor α (ER+) tumors, the molecular mechanisms for endocrine-resistant disease remain unresolved. Previously we demonstrated that expression of the RNA binding protein and N6-methyladenosine (m6A) reader HNRNPA2B1 (A2B1) is higher in LCC9 and LY2 tamoxifen (TAM)-resistant ERα breast cancer cells relative to parental TAM-sensitive MCF-7 cells. Here we report that A2B1 protein expression is higher in breast tumors than paired normal breast tissue. Modest stable overexpression of A2B1 in MCF-7 cells (MCF-7-A2B1 cells) resulted in TAM- and fulvestrant- resistance whereas knockdown of A2B1 in LCC9 and LY2 cells restored TAM and fulvestrant, endocrine-sensitivity. MCF-7-A2B1 cells gained hallmarks of TAM-resistant metastatic behavior: increased migration and invasion, clonogenicity, and soft agar colony size, which were attenuated by A2B1 knockdown in MCF-7-A2B1 and the TAM-resistant LCC9 and LY2 cells. MCF-7-A2B1, LCC9, and LY2 cells have a higher proportion of CD44+/CD24-/low cancer stem cells (CSC) compared to MCF-7 cells. MCF-7-A2B1 cells have increased ERα and reduced miR-222-3p that targets ERα. Like LCC9 cells, MCF-7-A2B1 have activated AKT and MAPK that depend on A2B1 expression and are growth inhibited by inhibitors of these pathways. These data support that targeting A2B1 could provide a complimentary therapeutic approach to reduce acquired endocrine resistance.
Collapse
Affiliation(s)
- Belinda J Petri
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Kellianne M Piell
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Gordon C South Whitt
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Ali E Wilt
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Claire C Poulton
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Norman L Lehman
- Department of Pathology and Laboratory Medicine, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Brian F Clem
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Matthew A Nystoriak
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Marcin Wysoczynski
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Carolyn M Klinge
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA.
| |
Collapse
|
30
|
Titratable Pharmacological Regulation of CAR T Cells Using Zinc Finger-Based Transcription Factors. Cancers (Basel) 2021; 13:cancers13194741. [PMID: 34638227 PMCID: PMC8507528 DOI: 10.3390/cancers13194741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/14/2021] [Accepted: 09/17/2021] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Chimeric antigen receptor (CAR) T cell therapy can be associated with substantial side effects primarily due to intense immune activation following treatment, or target antigen recognition on off-tumor tissue. Consequently, temporal and tunable control of CAR T cell activity is of major importance for the clinical translation of innovative CAR designs. This work demonstrates the transcriptional regulation of an anti-CD20 CAR in primary T cells using a drug inducible zinc finger-based transcription factor. The switch system enables titratable induction of CAR expression and CAR T cell effector function with the clinically relevant inducer drug tamoxifen and its metabolites both in vitro and in vivo, whereby CAR activity is strictly dependent on the presence of the inducer drug. The results obtained can readily be transferred to other CARs for which an improved control of expression is required. Abstract Chimeric antigen receptor (CAR) T cell therapy has emerged as an attractive strategy for cancer immunotherapy. Despite remarkable success for hematological malignancies, excessive activity and poor control of CAR T cells can result in severe adverse events requiring control strategies to improve safety. This work illustrates the feasibility of a zinc finger-based inducible switch system for transcriptional regulation of an anti-CD20 CAR in primary T cells providing small molecule-inducible control over therapeutic functions. We demonstrate time- and dose-dependent induction of anti-CD20 CAR expression and function with metabolites of the clinically-approved drug tamoxifen, and the absence of background CAR activity in the non-induced state. Inducible CAR T cells executed fine-tuned cytolytic activity against target cells both in vitro and in vivo, whereas CAR-related functions were lost upon drug discontinuation. This zinc finger-based transcriptional control system can be extended to other therapeutically important CARs, thus paving the way for safer cellular therapies.
Collapse
|
31
|
van der Lee M, Allard WG, Vossen RHAM, Baak-Pablo RF, Menafra R, Deiman BALM, Deenen MJ, Neven P, Johansson I, Gastaldello S, Ingelman-Sundberg M, Guchelaar HJ, Swen JJ, Anvar SY. Toward predicting CYP2D6-mediated variable drug response from CYP2D6 gene sequencing data. Sci Transl Med 2021; 13:13/603/eabf3637. [PMID: 34290055 DOI: 10.1126/scitranslmed.abf3637] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/11/2021] [Accepted: 06/14/2021] [Indexed: 12/14/2022]
Abstract
Pharmacogenomics is a key component of personalized medicine that promises safer and more effective drug treatment by individualizing drug choice and dose based on genetic profiles. In clinical practice, genetic biomarkers are used to categorize patients into *-alleles to predict CYP450 enzyme activity and adjust drug dosages accordingly. However, this approach leaves a large part of variability in drug response unexplained. Here, we present a proof-of-concept approach that uses continuous-scale (instead of categorical) assignments to predict enzyme activity. We used full CYP2D6 gene sequences obtained with long-read amplicon-based sequencing and cytochrome P450 (CYP) 2D6-mediated tamoxifen metabolism data from a prospective study of 561 patients with breast cancer to train a neural network. The model explained 79% of interindividual variability in CYP2D6 activity compared to 54% with the conventional *-allele approach, assigned enzyme activities to known alleles with previously reported effects, and predicted the activity of previously uncharacterized combinations of variants. The results were replicated in an independent cohort of tamoxifen-treated patients (model R 2 adjusted = 0.66 versus *-allele R 2 adjusted = 0.35) and a cohort of patients treated with the CYP2D6 substrate venlafaxine (model R 2 adjusted = 0.64 versus *-allele R 2 adjusted = 0.55). Human embryonic kidney cells were used to confirm the effect of five genetic variants on metabolism of the CYP2D6 substrate bufuralol in vitro. These results demonstrate the advantage of a continuous scale and a completely phased genotype for prediction of CYP2D6 enzyme activity and could potentially enable more accurate prediction of individual drug response.
Collapse
Affiliation(s)
- Maaike van der Lee
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands.,Leiden Network for Personalised Therapeutics, 2333 ZA Leiden, Netherlands
| | - William G Allard
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, Netherlands.,Leiden Genome Technology Center, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Rolf H A M Vossen
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, Netherlands.,Leiden Genome Technology Center, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Renée F Baak-Pablo
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Roberta Menafra
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, Netherlands.,Leiden Genome Technology Center, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | - Birgit A L M Deiman
- Clinical Laboratory, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands.,Department of Clinical Pharmacy, Catharina Hospital Eindhoven, 5623 EJ Eindhoven, Netherlands
| | | | - Inger Johansson
- Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum 5B, 171 77 Solna, Sweden
| | - Stefano Gastaldello
- Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum 5B, 171 77 Solna, Sweden
| | - Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Biomedicum 5B, 171 77 Solna, Sweden
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands.,Leiden Network for Personalised Therapeutics, 2333 ZA Leiden, Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands. .,Leiden Network for Personalised Therapeutics, 2333 ZA Leiden, Netherlands
| | - Seyed Yahya Anvar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, 2333 ZA Leiden, Netherlands. .,Leiden Network for Personalised Therapeutics, 2333 ZA Leiden, Netherlands.,Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, Netherlands.,Leiden Genome Technology Center, Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| |
Collapse
|
32
|
Fabian CJ. Tamoxifen: Will Less Equal More in Women with Precancerous Breast Disease? Clin Cancer Res 2021; 27:3510-3511. [PMID: 33926916 DOI: 10.1158/1078-0432.ccr-21-0729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/07/2021] [Accepted: 04/26/2021] [Indexed: 11/16/2022]
Abstract
Similar risk reduction but fewer side effects would predict more uptake and compliance with low (5 mg) versus full (20 mg) dose tamoxifen. Benefit with low dose is demonstrated for perimenopausal/postmenopausal women with intraepithelial neoplasia and high lesion Ki-67. Longer follow-up needed to determine benefit with low lesion Ki-67.See related article by DeCensi et al., p. 3576.
Collapse
Affiliation(s)
- Carol J Fabian
- Department of Internal Medicine, Division of Clinical Oncology, University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
33
|
Autophagy Triggers Tamoxifen Resistance in Human Breast Cancer Cells by Preventing Drug-Induced Lysosomal Damage. Cancers (Basel) 2021; 13:cancers13061252. [PMID: 33809171 PMCID: PMC7999102 DOI: 10.3390/cancers13061252] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Endocrine therapy with tamoxifen or other endocrine drugs represents the standard treatment for estrogen receptor-positive breast cancer. In spite of effectiveness of this therapy, onset of drug resistance worsens the prognosis of about 30% of patients. Autophagy has recently been proposed as a key player of drug resistance, but the underlying mechanisms are not completely understood. In this research, the authors investigate how autophagy triggers drug resistance in breast cancer cells. The results evidence that tamoxifen affects lysosome integrity, which suggests that this effect may contribute to the anticancer activity of this drug. Activation of autophagy and overexpression of iron-binding proteins synergize in protecting the lysosomal compartment, restraining drug effectiveness in breast cancer cells. According to these results, tamoxifen-resistant cells show an increased autophagic flux and overexpress iron-binding proteins. These findings indicate that screening for the level of iron-binding proteins may help to identify patients at risk for developing drug resistance. Abstract Endocrine resistance is a major complication during treatment of estrogen receptor-positive breast cancer. Although autophagy has recently gained increasing consideration among the causative factors, the link between autophagy and endocrine resistance remains elusive. Here, we investigate the autophagy-based mechanisms of tamoxifen resistance in MCF7 cells. Tamoxifen (Tam) triggers autophagy and affects the lysosomal compartment of MCF7 cells, such that activated autophagy supports disposal of tamoxifen-damaged lysosomes by lysophagy. MCF7 cells resistant to 5 µM tamoxifen (MCF7-TamR) have a higher autophagic flux and an enhanced resistance to Tam-induced lysosomal alterations compared to parental cells, which suggests a correlation between the two events. MCF7-TamR cells overexpress messenger RNAs (mRNAs) for metallothionein 2A and ferritin heavy chain, and they are re-sensitized to Tam by inhibition of autophagy. Overexpressing these proteins in parental MCF7 cells protects lysosomes from Tam-induced damage and preserves viability, while inhibiting autophagy abrogates lysosome protection. Consistently, we also demonstrate that other breast cancer cells that overexpress selected mRNAs encoding iron-binding proteins are less sensitive to Tam-induced lysosomal damage when autophagy is activated. Collectively, our data demonstrate that autophagy triggers Tam resistance in breast cancer cells by favoring the lysosomal relocation of overexpressed factors that restrain tamoxifen-induced lysosomal damage.
Collapse
|
34
|
Sula A, Hollingworth D, Ng LCT, Larmore M, DeCaen PG, Wallace BA. A tamoxifen receptor within a voltage-gated sodium channel. Mol Cell 2021; 81:1160-1169.e5. [PMID: 33503406 PMCID: PMC7980221 DOI: 10.1016/j.molcel.2020.12.048] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 08/24/2020] [Accepted: 12/14/2020] [Indexed: 12/22/2022]
Abstract
Voltage-gated sodium channels are targets for many analgesic and antiepileptic drugs whose therapeutic mechanisms and binding sites have been well characterized. We describe the identification of a previously unidentified receptor site within the NavMs voltage-gated sodium channel. Tamoxifen, an estrogen receptor modulator, and its primary and secondary metabolic products bind at the intracellular exit of the channel, which is a site that is distinct from other previously characterized sodium channel drug sites. These compounds inhibit NavMs and human sodium channels with similar potencies and prevent sodium conductance by delaying channel recovery from the inactivated state. This study therefore not only describes the structure and pharmacology of a site that could be leveraged for the development of new drugs for the treatment of sodium channelopathies but may also have important implications for off-target health effects of this widely used therapeutic drug.
Collapse
Affiliation(s)
- Altin Sula
- Institute of Structural and Molecular Biology, Birkbeck College, University of London, London WC1E 7HX, UK
| | - David Hollingworth
- Institute of Structural and Molecular Biology, Birkbeck College, University of London, London WC1E 7HX, UK
| | - Leo C T Ng
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Megan Larmore
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Paul G DeCaen
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - B A Wallace
- Institute of Structural and Molecular Biology, Birkbeck College, University of London, London WC1E 7HX, UK.
| |
Collapse
|
35
|
Slanař O, Hronová K, Bartošová O, Šíma M. Recent advances in the personalized treatment of estrogen receptor-positive breast cancer with tamoxifen: a focus on pharmacogenomics. Expert Opin Drug Metab Toxicol 2020; 17:307-321. [PMID: 33320718 DOI: 10.1080/17425255.2021.1865310] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: Tamoxifen is still an important drug in hormone-dependent breast cancer therapy. Personalization of its clinical use beyond hormone receptor positivity could improve the substantial variability of the treatment response.Areas covered: The overview of the current evidence for the treatment personalization using therapeutic drug monitoring, or using genetic biomarkers including CYP2D6 is provided. Although many studies focused on the PK aspects or the impact of CYP2D6 variability the translation into clinical routine is not clearly defined due to the inconsistent clinical outcome data.Expert opinion: We believe that at least the main candidate factors, i.e. CYP2D6 polymorphism, CYP2D6 inhibition, endoxifen serum levels may become important predictors of clinical relevance for tamoxifen treatment personalization in the future. To achieve this aim, however, further research should take into consideration more precise characterization of the disease, epigenetic factors and also utilize an appropriately powered multifactorial approach instead of a single gene evaluating studies.
Collapse
Affiliation(s)
- Ondřej Slanař
- Department of Pharmacology, Charles University and General University Hospital, Prague, Czech Republic
| | - Karolína Hronová
- Department of Pharmacology, Charles University and General University Hospital, Prague, Czech Republic
| | - Olga Bartošová
- Department of Pharmacology, Charles University and General University Hospital, Prague, Czech Republic
| | - Martin Šíma
- Department of Pharmacology, Charles University and General University Hospital, Prague, Czech Republic
| |
Collapse
|
36
|
Anlaş AA, Nelson CM. Soft Microenvironments Induce Chemoresistance by Increasing Autophagy Downstream of Integrin-Linked Kinase. Cancer Res 2020; 80:4103-4113. [PMID: 33008805 PMCID: PMC7534696 DOI: 10.1158/0008-5472.can-19-4021] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/28/2020] [Accepted: 07/22/2020] [Indexed: 01/03/2023]
Abstract
Breast cancer relapse can develop over the course of years as a result of dormant cancer cells that disseminate to secondary sites. These dormant cells are often resistant to conventional hormone and chemotherapy. Although recurrence is the main cause of death from cancer, microenvironmental factors that may influence resistance to therapy and duration of dormancy are largely unknown. Breast cancer relapse is often detected in tissues that are softer than the normal mammary gland or the primary breast tumor, such as bone marrow, brain, and lung. We therefore explored how stiffness of the microenvironment at secondary sites regulates tumor dormancy and the response of breast cancer cells to hormone and chemotherapy. In soft microenvironments reminiscent of metastatic sites, breast cancer cells were more resistant to the estrogen receptor modulator tamoxifen as a result of increased autophagy and decreased expression of estrogen receptor-α. Consistently, pharmacologic inhibition or genetic downregulation of autophagy increased the response of breast cancer cells to tamoxifen on soft substrata. In addition, autophagy was decreased downstream of integrin-linked kinase on stiff substrata. Altogether, our data show that tissue mechanics regulates therapeutic outcome and long-term survival of breast cancer cells by influencing autophagy. SIGNIFICANCE: These findings characterize the persistence of dormant cells at metastatic sites, where soft microenvironments downregulate estrogen receptor expression and upregulate autophagy, thereby promoting therapy resistance in breast cancer cells. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/19/4103/F1.large.jpg.
Collapse
Affiliation(s)
- Alişya A Anlaş
- Department of Chemical and Biological Engineering, Princeton, New Jersey
| | - Celeste M Nelson
- Department of Chemical and Biological Engineering, Princeton, New Jersey.
- Department of Molecular Biology Princeton University, Princeton, New Jersey
| |
Collapse
|
37
|
Molina L, Bustamante F, Ortloff A, Ramos I, Ehrenfeld P, Figueroa CD. Continuous Exposure of Breast Cancer Cells to Tamoxifen Upregulates GPER-1 and Increases Cell Proliferation. Front Endocrinol (Lausanne) 2020; 11:563165. [PMID: 33117280 PMCID: PMC7561417 DOI: 10.3389/fendo.2020.563165] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/08/2020] [Indexed: 12/13/2022] Open
Abstract
GPER-1 is a novel membrane sited G protein-coupled estrogen receptor. Clinical studies have shown that patients suffering an estrogen receptor α (ERα)/GPER-1 positive, breast cancer have a lower survival rate than those who have developed ERα-positive/GPER-1 negative tumors. Moreover, absence of GPER-1 improves the prognosis of patients treated with tamoxifen, the most used selective estrogen receptor modulator to treat ERα-positive breast cancer. MCF-7 breast cancer cells were continuously treated with 1,000 nM tamoxifen for 7 days to investigate its effect on GPER-1 protein expression, cell proliferation and intracellular [Ca2+]i mobilization, a key signaling pathway. Breast cancer cells continuously treated with tamoxifen, exhibited a robust [Ca2+]i mobilization after stimulation with 1,000 nM tamoxifen, a response that was blunted by preincubation of cells with G15, a commercial GPER-1 antagonist. Continuously treated cells also displayed a high [Ca2+]i mobilization in response to a commercial GPER-1 agonist (G1) and to estrogen, in a magnitude that doubled the response observed in untreated cells and was almost completely abolished by G15. Proliferation of cells continuously treated with tamoxifen and stimulated with 2,000 nM tamoxifen, was also higher than that observed in untreated cells in a degree that was approximately 90% attributable to GPER-1. Finally, prolonged tamoxifen treatment did not increase ERα expression, but did overexpress the kinin B1 receptor, another GPCR, which we have previously shown is highly expressed in breast tumors and increases proliferation of breast cancer cells. Although we cannot fully extrapolate the results obtained in vitro to the patients, our results shed some light on the occurrence of drug resistance in breast cancer patients who are ERα/GPER-1 positive, have been treated with tamoxifen and display low survival rate. Overexpression of kinin B1 receptor may explain the increased proliferative response observed in breast tumors under continuous treatment with tamoxifen.
Collapse
Affiliation(s)
- Luis Molina
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Puerto Montt, Chile
| | - Felipe Bustamante
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Universidad Austral de Chile, Valdivia, Chile
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Alexander Ortloff
- Departamento de Ciencias Veterinarias y Salud Pública, Facultad de Recursos Naturales, Universidad Católica de Temuco, Temuco, Chile
| | - Iraidi Ramos
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Universidad Austral de Chile, Valdivia, Chile
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Pamela Ehrenfeld
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Universidad Austral de Chile, Valdivia, Chile
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Carlos D. Figueroa
- Laboratory of Cellular Pathology, Institute of Anatomy, Histology and Pathology, Universidad Austral de Chile, Valdivia, Chile
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Universidad Austral de Chile, Valdivia, Chile
| |
Collapse
|
38
|
Zhang X, de Oliveira Andrade F, Zhang H, Cruz I, Clarke R, Gaur P, Verma V, Hilakivi-Clarke L. Maternal obesity increases offspring's mammary cancer recurrence and impairs tumor immune response. Endocr Relat Cancer 2020; 27:469-482. [PMID: 32580156 PMCID: PMC7424355 DOI: 10.1530/erc-20-0065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
Abstract
Over 50% of women at a childbearing age in the United States are overweight or obese, and this can adversely affect their offspring. We studied if maternal obesity-inducing high fat diet (HFD) not only increases offspring's mammary cancer risk but also impairs response to antiestrogen tamoxifen. Female rat offspring of HFD and control diet-fed dams, in which estrogen receptor-positive (ER+) mammary tumors were induced with the carcinogen 7,12-dimethylbenz[a]anthracene (DMBA), exhibited similar initial responses to antiestrogen tamoxifen. However, after tamoxifen therapy was completed, almost all (91%) tumors recurred in HFD offspring, compared with only 29% in control offspring. The increase in local mammary tumor recurrence in HFD offspring was linked to an increase in the markers of immunosuppression (Il17f, Tgfβ1, VEGFR2) in the tumor microenvironment (TME). Protein and mRNA levels of the major histocompatibility complex II (MHC-II), but not MHC-I, were reduced in the recurring DMBA tumors of HFD offspring. Further, infiltration of CD8+ effector T cells and granzyme B+ (GZMB+) cells were lower in their recurring tumors. To determine if maternal HFD can pre-program similar changes in the TME of allografted E0771 mammary tumors in offspring of syngeneic mice, flow cytometry analysis was performed. E0771 mammary tumor growth was significantly accelerated in the HFD offspring, and a reduction in the numbers of GZMB and non-significant reduction of interferon γ (IFNγ) secreting CD8+ T cells in the TME was seen. Thus, consumption of a HFD during pregnancy increases susceptibility of the female rat and mouse offspring to tumor immune suppression and mammary tumor growth and recurrence.
Collapse
Affiliation(s)
| | | | | | | | - Robert Clarke
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Pankaj Gaur
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Vivek Verma
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Leena Hilakivi-Clarke
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
- Correspondence should be addressed to L Hilakivi-Clarke:
| |
Collapse
|
39
|
Mohr CJ, Schroth W, Mürdter TE, Gross D, Maier S, Stegen B, Dragoi A, Steudel FA, Stehling S, Hoppe R, Madden S, Ruth P, Huber SM, Brauch H, Lukowski R. Subunits of BK channels promote breast cancer development and modulate responses to endocrine treatment in preclinical models. Br J Pharmacol 2020; 179:2906-2924. [PMID: 32468618 DOI: 10.1111/bph.15147] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 03/20/2020] [Accepted: 05/13/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Pore-forming α subunits of the voltage- and Ca2+ -activated K+ channel with large conductance (BKα) promote malignant phenotypes of breast tumour cells. Auxiliary subunits such as the leucine-rich repeat containing 26 (LRRC26) protein, also termed BKγ1, may be required to permit activation of BK currents at a depolarized resting membrane potential that frequently occur in non-excitable tumour cells. EXPERIMENTAL APPROACH Anti-tumour effects of BKα loss were investigated in breast tumour-bearing MMTV-PyMT transgenic BKα knockout (KO) mice, primary MMTV-PyMT cell cultures, and in a syngeneic transplantation model of breast cancer derived from these cells. The therapeutic relevance of BK channels in the context of endocrine treatment was assessed in human breast cancer cell lines expressing either low (MCF-7) or high (MDA-MB-453) levels of BKα and BKγ1, as well as in BKα-negative MDA-MB-157. KEY RESULTS BKα promoted breast cancer onset and overall survival in preclinical models. Conversely, lack of BKα and/or knockdown of BKγ1 attenuated proliferation of murine and human breast cancer cells in vitro. At low concentrations, tamoxifen and its major active metabolites stimulated proliferation of BKα/γ1-positive breast cancer cells, independent of the genomic signalling controlled by the oestrogen receptor. Finally, tamoxifen increased the relative survival time of BKα KO but not of wild-type tumour cell recipient mice. CONCLUSION AND IMPLICATIONS Breast cancer initiation, progression, and tamoxifen sensitivity depend on functional BK channels thereby providing a rationale for the future exploration of the oncogenic actions of BK channels in clinical outcomes with anti-oestrogen therapy.
Collapse
Affiliation(s)
- Corinna J Mohr
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany.,Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart and University of Tuebingen, Germany
| | - Werner Schroth
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart and University of Tuebingen, Germany
| | - Thomas E Mürdter
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart and University of Tuebingen, Germany
| | - Dominic Gross
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany
| | - Selina Maier
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany.,Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart and University of Tuebingen, Germany
| | - Benjamin Stegen
- Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - Alice Dragoi
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany
| | - Friederike A Steudel
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany
| | - Severine Stehling
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany
| | - Reiner Hoppe
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart and University of Tuebingen, Germany
| | - Stephen Madden
- RCSI Division of Population Health Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany
| | - Stephan M Huber
- Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - Hiltrud Brauch
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart and University of Tuebingen, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,iFIT-Cluster of Excellence, University of Tuebingen, Tuebingen, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
40
|
White‐Gilbertson S, Lu P, Jones CM, Chiodini S, Hurley D, Das A, Delaney JR, Norris JS, Voelkel‐Johnson C. Tamoxifen is a candidate first-in-class inhibitor of acid ceramidase that reduces amitotic division in polyploid giant cancer cells-Unrecognized players in tumorigenesis. Cancer Med 2020; 9:3142-3152. [PMID: 32135040 PMCID: PMC7196070 DOI: 10.1002/cam4.2960] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/18/2022] Open
Abstract
Polyploid giant cancer cells (PGCC) represent a poorly understood, small subpopulation of tumor cells that are increasingly being recognized for their critical role in therapy resistance, metastasis, and cancer recurrence. PGCC have the potential to generate progeny through primitive or cleavage-like division, which allows them to evade antimitotic insults. We recently demonstrated that the sphingolipid enzyme acid ceramidase (ASAH1) is required for this process. Since specific ASAH1 inhibitors are not clinically available, we investigated whether tamoxifen, which interferes with ASAH1 function via off-target effects, has a potential clinical benefit independent of estrogen signaling. Our results show that tamoxifen inhibits generation of PGCC offspring in prostate cancer, glioblastoma, and melanoma cells. Analysis of two state-level cancer registries revealed that tamoxifen improves survival outcomes for second, nonbreast cancers that develop in women with early stage breast cancer. Our results suggest that tamoxifen may have a clinical benefit in a variety of cancers that is independent of estrogen signaling and could be due to its inhibition of acid ceramidase. Thus the distinct application of tamoxifen as potentially a first-in-class therapeutic that inhibits the generation of PGCC offspring should be considered in future clinical trials.
Collapse
Affiliation(s)
- Shai White‐Gilbertson
- Department of Microbiology & ImmunologyMedical University of South CarolinaCharlestonSCUSA
| | - Ping Lu
- Department of Microbiology & ImmunologyMedical University of South CarolinaCharlestonSCUSA
| | - Christian M. Jones
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSCUSA
| | | | - Deborah Hurley
- South Carolina Central Cancer RegistrySCDHECColumbiaSCUSA
| | - Arabinda Das
- Department of NeuroscienceMedical University of South CarolinaCharlestonSCUSA
| | - Joe R. Delaney
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSCUSA
| | - James S. Norris
- Department of Microbiology & ImmunologyMedical University of South CarolinaCharlestonSCUSA
| | - Christina Voelkel‐Johnson
- Department of Microbiology & ImmunologyMedical University of South CarolinaCharlestonSCUSA
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSCUSA
| |
Collapse
|
41
|
Klopp-Schulze L, Mueller-Schoell A, Neven P, Koolen SLW, Mathijssen RHJ, Joerger M, Kloft C. Integrated Data Analysis of Six Clinical Studies Points Toward Model-Informed Precision Dosing of Tamoxifen. Front Pharmacol 2020; 11:283. [PMID: 32296331 PMCID: PMC7136483 DOI: 10.3389/fphar.2020.00283] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/27/2020] [Indexed: 12/16/2022] Open
Abstract
Introduction At tamoxifen standard dosing, ∼20% of breast cancer patients do not reach proposed target endoxifen concentrations >5.97 ng/mL. Thus, better understanding the large interindividual variability in tamoxifen pharmacokinetics (PK) is crucial. By applying non-linear mixed-effects (NLME) modeling to a pooled ‘real-world’ clinical PK database, we aimed to (i) dissect several levels of variability and identify factors predictive for endoxifen exposure and (ii) assess different tamoxifen dosing strategies for their potential to increase the number of patients reaching target endoxifen concentrations. Methods Tamoxifen and endoxifen concentrations with genetic and demographic data of 468 breast cancer patients from six reported studies were used to develop a NLME parent-metabolite PK model. Different levels of variability on model parameters or measurements were investigated and the impact of covariates thereupon explored. The model was subsequently applied in a simulation-based comparison of three dosing strategies with increasing degree of dose individualization for a large virtual breast cancer population. Interindividual variability of endoxifen concentrations and the fraction of patients at risk for not reaching target concentrations were assessed for each dosing strategy. Results and Conclusions The integrated NLME model enabled to differentiate and quantify four levels of variability (interstudy, interindividual, interoccasion, and intraindividual). Strong influential factors, i.e., CYP2D6 activity score, drug–drug interactions with CYP3A and CYP2D6 inducers/inhibitors and age, were reliably identified, reducing interoccasion variability to <20% CV. Yet, unexplained interindividual variability in endoxifen formation remained large (47.2% CV). Hence, therapeutic drug monitoring seems promising for achieving endoxifen target concentrations. Three tamoxifen dosing strategies [standard dosing (20 mg QD), CYP2D6-guided dosing (20, 40, and 60 mg QD) and individual model-informed precision dosing (MIPD)] using three therapeutic drug monitoring samples (5–120 mg QD) were compared, leveraging the model. The proportion of patients at risk for not reaching target concentrations was 22.2% in standard dosing, 16.0% in CYP2D6-guided dosing and 7.19% in MIPD. While in CYP2D6-guided- and standard dosing interindividual variability in endoxifen concentrations was high (64.0% CV and 68.1% CV, respectively), it was considerably reduced in MIPD (24.0% CV). Hence, MIPD demonstrated to be the most promising strategy for achieving target endoxifen concentrations.
Collapse
Affiliation(s)
- Lena Klopp-Schulze
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Free University of Berlin, Berlin, Germany
| | - Anna Mueller-Schoell
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Free University of Berlin, Berlin, Germany.,Graduate Research Training Program PharMetrX, Berlin, Germany
| | - Patrick Neven
- Vesalius Research Center, University Hospitals Leuven, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Stijn L W Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Ron H J Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
| | - Markus Joerger
- Department of Medical Oncology and Hematology, Cantonal Hospital, St., Gallen, Switzerland
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Free University of Berlin, Berlin, Germany
| |
Collapse
|
42
|
Weinstock A, Gallego-Delgado J, Gomes C, Sherman J, Nikain C, Gonzalez S, Fisher E, Rodriguez A. Tamoxifen activity against Plasmodium in vitro and in mice. Malar J 2019; 18:378. [PMID: 31775753 PMCID: PMC6882195 DOI: 10.1186/s12936-019-3012-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/16/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Tamoxifen is an oestrogen receptor modulator that is widely used for the treatment of early stage breast cancer and reduction of recurrences. Tamoxifen is also used as a powerful research tool for controlling gene expression in the context of the Cre/loxP site-specific recombination system in conditional mutant mice. METHODS To determine whether the administration of tamoxifen affects Plasmodium growth and/or disease outcome in malaria, in vitro studies assessing the effect of tamoxifen and its active metabolite 4-hydroxytamoxifen on Plasmodium falciparum blood stages were performed. Tamoxifen effects were also evaluated in vivo treating C57/B6 mice infected with Plasmodium berghei (ANKA strain), which is the standard animal model for the study of cerebral malaria. RESULTS Tamoxifen and its active metabolite, 4-hydroxytamoxifen, show activity in vitro against P. falciparum (16.7 to 5.8 µM IC50, respectively). This activity was also confirmed in tamoxifen-treated mice infected with P. berghei, which show lower levels of parasitaemia and do not develop signs of cerebral malaria, compared to control mice. Mice treated with tamoxifen for 1 week and left untreated for an additional week before infection showed similar parasitaemia levels and signs of cerebral malaria as control untreated mice. CONCLUSIONS Tamoxifen and its active metabolite, 4-hydroxytamoxifen, have significant activity against the human parasite P. falciparum in vitro and the rodent parasite P. berghei in vivo. This activity may be useful for prevention of malaria in patients taking this drug chronically, but also represents a major problem for scientists using the conditional mutagenic Cre/LoxP system in the setting of rodent malaria. Allowing mice to clear tamoxifen before starting a Plasmodium infection allows the use the Cre/LoxP conditional mutagenic system to investigate gene function in specific tissues.
Collapse
Affiliation(s)
- Ada Weinstock
- Departments of Medicine (Cardiology) and Cell Biology, and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Julio Gallego-Delgado
- Department of Biological Sciences, Lehman College, City University of New York, Bronx, New York, NY, 10468, USA.
- Ph.D. Program in Biology, The Graduate Center, The City University of New York, New York, NY, 10016, USA.
- Department of Microbiology, New York University School of Medicine, New York, NY, 10016, USA.
| | - Cláudia Gomes
- Department of Microbiology, New York University School of Medicine, New York, NY, 10016, USA
| | - Julian Sherman
- Department of Microbiology, New York University School of Medicine, New York, NY, 10016, USA
| | - Cyrus Nikain
- Departments of Medicine (Cardiology) and Cell Biology, and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Sandra Gonzalez
- Department of Microbiology, New York University School of Medicine, New York, NY, 10016, USA
| | - Edward Fisher
- Departments of Medicine (Cardiology) and Cell Biology, and the Marc and Ruti Bell Program in Vascular Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Ana Rodriguez
- Department of Microbiology, New York University School of Medicine, New York, NY, 10016, USA
| |
Collapse
|
43
|
Tomková V, Sandoval-Acuña C, Torrealba N, Truksa J. Mitochondrial fragmentation, elevated mitochondrial superoxide and respiratory supercomplexes disassembly is connected with the tamoxifen-resistant phenotype of breast cancer cells. Free Radic Biol Med 2019; 143:510-521. [PMID: 31494243 DOI: 10.1016/j.freeradbiomed.2019.09.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/04/2019] [Accepted: 09/04/2019] [Indexed: 12/22/2022]
Abstract
Tamoxifen resistance remains a clinical obstacle in the treatment of hormone sensitive breast cancer. It has been reported that tamoxifen is able to target respiratory complex I within mitochondria. Therefore, we established two tamoxifen-resistant cell lines, MCF7 Tam5R and T47D Tam5R resistant to 5 μM tamoxifen and investigated whether tamoxifen-resistant cells exhibit mitochondrial changes which could help them survive the treatment. The function of mitochondria in this experimental model was evaluated in detail by studying i) the composition and activity of mitochondrial respiratory complexes; ii) respiration and glycolytic status; iii) mitochondrial distribution, dynamics and reactive oxygen species production. We show that Tam5R cells exhibit a significant decrease in mitochondrial respiration, low abundance of assembled mitochondrial respiratory supercomplexes, a more fragmented mitochondrial network connected with DRP1 Ser637 phosphorylation, higher glycolysis and sensitivity to 2-deoxyglucose. Tam5R cells also produce significantly higher levels of mitochondrial superoxide but at the same time increase their antioxidant defense (CAT, SOD2) through upregulation of SIRT3 and show phosphorylation of AMPK at Ser 485/491. Importantly, MCF7 ρ0 cells lacking functional mitochondria exhibit a markedly higher resistance to tamoxifen, supporting the role of mitochondria in tamoxifen resistance. We propose that reduced mitochondrial function and higher level of reactive oxygen species within mitochondria in concert with metabolic adaptations contribute to the phenotype of tamoxifen resistance.
Collapse
Affiliation(s)
- Veronika Tomková
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | | | - Natalia Torrealba
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Jaroslav Truksa
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic.
| |
Collapse
|
44
|
Hasan M, Marzouk MA, Adhikari S, Wright TD, Miller BP, Matossian MD, Elliott S, Wright M, Alzoubi M, Collins-Burow BM, Burow ME, Holzgrabe U, Zlotos DP, Stratford RE, Witt-Enderby PA. Pharmacological, Mechanistic, and Pharmacokinetic Assessment of Novel Melatonin-Tamoxifen Drug Conjugates as Breast Cancer Drugs. Mol Pharmacol 2019; 96:272-296. [PMID: 31221824 PMCID: PMC6666385 DOI: 10.1124/mol.119.116202] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/07/2019] [Indexed: 02/05/2023] Open
Abstract
Tamoxifen is used to prevent and treat estrogen receptor-positive (ER+) breast cancer (BC); however, its chronic use can increase uterine cancer risk and induce tamoxifen resistance. Novel melatonin-tamoxifen drug conjugates may be promising to treat BC and may help offset the adverse effects of tamoxifen usage alone due to the presence of melatonin. We synthesized and screened five drug conjugates (C2, C4, C5, C9, and C15 linked) for their effects on BC cell (MCF-7, tamoxifen-resistant MCF-7, mouse mammary carcinoma, MDA-MB-231, and BT-549) viability, migration, and binding affinity to melatonin receptor 1 (MT1R) and estrogen receptor 1 (ESR1). C4 and C5 demonstrated the most favorable pharmacological characteristics with respect to binding profiles (affinity for ESR1 and MT1R) and their potency/efficacy to inhibit BC cell viability and migration in four phenotypically diverse invasive ductal BC cell lines. C4 and C5 were further assessed for their actions against tamoxifen-resistant MCF-7 cells and a patient-derived xenograft triple-negative BC cell line (TU-BcX-4IC) and for their mechanisms of action using selective mitogen-activated protein kinase kinase MEK1/2, MEK5, and phosphoinositide 3-kinase (PI3K) inhibitors. C4 and C5 inhibited tamoxifen-resistant MCF-7 cells with equal potency (IC50 = 4-8 μM) and efficacy (∼90% inhibition of viability and migration) but demonstrated increased potency (IC50 = 80-211 μM) and efficacy (∼140% inhibition) to inhibit migration versus cell viability (IC50 = 181-304 mM; efficacy ∼80% inhibition) in TU-BcX-4IC cells. Unique pharmacokinetic profiles were observed, with C4 having greater bioavailability than C5. Further assessment of C4 and C5 demonstrates that they create novel pharmacophores within each BC cell that is context specific and involves MEK1/2/pERK1/2, MEK5/pERK5, PI3K, and nuclear factor κB. These melatonin-tamoxifen drug conjugates show promise as novel anticancer drugs and further preclinical and clinical evaluation is warranted.
Collapse
Affiliation(s)
- Mahmud Hasan
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Mohamed Akmal Marzouk
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Saugat Adhikari
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Thomas D Wright
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Benton P Miller
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Margarite D Matossian
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Steven Elliott
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Maryl Wright
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Madlin Alzoubi
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Bridgette M Collins-Burow
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Matthew E Burow
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Ulrike Holzgrabe
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Darius P Zlotos
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Robert E Stratford
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| | - Paula A Witt-Enderby
- Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (M.H., T.D.W., B.P.M., P.A.W.-E.); Department of Pharmaceutical Chemistry, German University in Cairo, New Cairo City, Cairo, Egypt (M.A.M., D.P.Z.); Purdue University, West Lafayette, Indiana (S.A.); Section of Hematology and Medical Oncology, Department of Medicine, Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana (M.D.M., S.E., M.W., M.A., B.M.C.-B., M.E.B.); Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, Wuerzburg, Germany (U.H.); Indiana University School of Medicine, Indianapolis, Indiana (R.E.S.); and Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania (P.A.W.-E.)
| |
Collapse
|
45
|
Fabian CJ. Will a Low-Dose Option Improve Uptake of Tamoxifen for Breast Cancer Risk Reduction? J Clin Oncol 2019; 37:1595-1597. [DOI: 10.1200/jco.19.00656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
46
|
Mina SG, Alaybeyoglu B, Murphy WL, Thomson JA, Stokes CL, Cirit M. Assessment of Drug-Induced Toxicity Biomarkers in the Brain Microphysiological System (MPS) Using Targeted and Untargeted Molecular Profiling. Front Big Data 2019; 2:23. [PMID: 33693346 PMCID: PMC7931859 DOI: 10.3389/fdata.2019.00023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/05/2019] [Indexed: 12/19/2022] Open
Abstract
Early assessment of adverse drug effects in humans is critical to avoid long-lasting harm. However, current approaches for early detection of adverse effects still lack predictive and organ-specific biomarkers to evaluate undesired responses in humans. Microphysiological systems (MPSs) are in vitro representations of human tissues and provide organ-specific translational insights for physiological processes. In this study, a brain MPS was utilized to assess molecular signatures of neurotoxic and non-neurotoxic compounds using targeted and untargeted molecular approaches. The brain MPS comprising of human embryonic stem (ES) cell-derived neural progenitor cells seeded on three-dimensional (3D), chemically defined, polyethylene glycol hydrogels was treated with the neurotoxic drug, bortezomib and the non-neurotoxic drug, tamoxifen over 14-days. Possible toxic effects were monitored with human N-acetylaspartic acid (NAA) kinetics, which correlates the neuronal function/health and DJ-1/PARK7, an oxidative stress biomarker. Changes in NAA levels were observed as early as 2-days post-bortezomib treatment, while onset detection of oxidative stress (DJ-1) was delayed until 4-days post-treatment. Separately, the untargeted extracellular metabolomics approach revealed distinct fingerprints 2-days post-bortezomib treatment as perturbations in cysteine and glycerophospholipid metabolic pathways. These results suggest accumulation of reactive oxygen species associated with oxidative stress, and disruption of membrane structure and integrity. The NAA response was strongly correlated with changes in a subset of the detected metabolites at the same time point 2-days post-treatment. Moreover, these metabolite changes correlated strongly with DJ-1 levels measured at the later time point (4-days post-treatment). This suggests that early cellular metabolic dysfunction leads to later DJ-1 leakage and cell death, and that early measurement of this subset of metabolites could predict the later occurrence of cell death. While the approach demonstrated here provides an individual case study for proof of concept, we suggest that this approach can be extended for preclinical toxicity screening and biomarker discovery studies.
Collapse
Affiliation(s)
- Sara G. Mina
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Begum Alaybeyoglu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - William L. Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - James A. Thomson
- Regenerative Biology, The Morgridge Institute for Research, Madison, WI, United States
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI, United States
| | | | - Murat Cirit
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
47
|
Liu J, Shelkar GP, Zhao F, Clausen RP, Dravid SM. Modulation of burst firing of neurons in nucleus reticularis of the thalamus by GluN2C-containing NMDA receptors. Mol Pharmacol 2019; 96:mol.119.116780. [PMID: 31160332 PMCID: PMC6620419 DOI: 10.1124/mol.119.116780] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/17/2019] [Accepted: 05/24/2019] [Indexed: 12/11/2022] Open
Abstract
The GluN2C subunit of the NMDA receptor is enriched in the neurons in nucleus reticularis of the thalamus (nRT), but its role in regulating their function is not well understood. We found that deletion of GluN2C subunit did not affect spike frequency in response to depolarizing current injection or hyperpolarization-induced rebound burst firing of nRT neurons. D-cycloserine or CIQ (GluN2C/GluN2D positive allosteric modulator) did not affect the depolarization-induced spike frequency in nRT neurons. A newly identified highly potent and efficacious co-agonist of GluN1/GluN2C NMDA receptors, AICP, was found to reduce the spike frequency and burst firing of nRT neurons in wildtype but not GluN2C knockout. This effect was potentially due to facilitation of GluN2C-containing receptors because inhibition of NMDA receptors by AP5 did not affect spike frequency in nRT neurons. We evaluated the effect of intracerebroventricular injection of AICP. AICP did not affect basal locomotion or prepulse inhibition but facilitated MK-801-induced hyperlocomotion. This effect was observed in wildtype but not in GluN2C knockout mice demonstrating that AICP produces GluN2C-selective effects in vivo Using a chemogenetic approach we examined the role of nRT in this behavioral effect. Gq or Gi coupled DREADDs were selectively expressed in nRT neurons using cre-dependent viral vectors and PV-Cre mouse line. We found that similar to AICP effect, activation of Gq but not Gi coupled DREADD facilitated MK-801-induced hyperlocomotion. Together, these results identify a unique role of GluN2C-containing receptors in the regulation of nRT neurons and suggest GluN2C-selective in vivo targeting of NMDA receptors by AICP. SIGNIFICANCE STATEMENT: The nucleus reticularis of the thalamus composed of GABAergic neurons is termed as guardian of the gateway and is an important regulator of corticothalamic communication which may be impaired in autism, non-convulsive seizures and other conditions. We found that strong facilitation of tonic activity of GluN2C subtype of NMDA receptors using AICP, a newly identified glycine-site agonist of NMDA receptors, modulates the function of reticular thalamus neurons. AICP was also able to produce GluN2C-dependent behavioral effects in vivo. Together, these finding identify a novel mechanism and a pharmacological tool to modulate activity of reticular thalamic neurons in disease states.
Collapse
|
48
|
Hruska CB, Hunt KN, Conners AL, Geske JR, Brandt KR, Degnim AC, Vachon CM, O'Connor MK, Rhodes DJ. Impact of short-term low-dose tamoxifen on molecular breast imaging background parenchymal uptake: a pilot study. Breast Cancer Res 2019; 21:38. [PMID: 30850011 PMCID: PMC6408779 DOI: 10.1186/s13058-019-1120-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 02/14/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND High background parenchymal uptake (BPU) on molecular breast imaging (MBI) has been identified as a breast cancer risk factor. We explored the feasibility of offering a short-term intervention of low-dose oral tamoxifen to women with high BPU and examined whether this intervention would reduce BPU. METHODS Women with a history of high BPU and no breast cancer history were invited to the study. Participants had an MBI exam, followed by 30 days of low-dose oral tamoxifen at either 5 mg or 10 mg/day, and a post-tamoxifen MBI exam. BPU on pre- and post-tamoxifen MBI exams was quantitatively assessed as the ratio of average counts in breast fibroglandular tissue vs. average counts in subcutaneous fat. Pre-tamoxifen and post-tamoxifen BPU were compared with paired t tests. RESULTS Of 47 women invited, 22 enrolled and 21 completed the study (10 taking 5 mg tamoxifen, 11 taking 10 mg tamoxifen). Mean age was 47.7 years (range 41-56 years). After 30 days low-dose tamoxifen, 8 of 21 women (38%) showed a decline in BPU, defined as a decrease from the pre-tamoxifen MBI of at least 15%; 11 of 21 (52%) had no change in BPU (within ± 15%); 2 of 21 (10%) had an increase in BPU of greater than 15%. Overall, the average post-tamoxifen BPU was not significantly different from pre-tamoxifen BPU (1.34 post vs. 1.43 pre, p = 0.11). However, among women taking 10 mg tamoxifen, 5 of 11 (45%) showed a decline in BPU; average BPU was 1.19 post-tamoxifen vs. 1.34 pre-tamoxifen (p = 0.005). In women taking 5 mg tamoxifen, 2 of 10 (20%) showed a decline in BPU; average BPU was 1.51 post-tamoxifen vs.1.53 pre-tamoxifen (p = 0.99). CONCLUSIONS Short-term intervention with low-dose tamoxifen may reduce high BPU on MBI for some patients. Our preliminary findings suggest that 10 mg tamoxifen per day may be more effective than 5 mg for inducing declines in BPU within 30 days. Given the variability in BPU response to tamoxifen observed among study participants, future study is warranted to determine if BPU response could predict the effectiveness of tamoxifen for breast cancer risk reduction within an individual. TRIAL REGISTRATION ClinicalTrials.gov NCT02979301 . Registered 01 December 2016.
Collapse
Affiliation(s)
- Carrie B Hruska
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Katie N Hunt
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Amy Lynn Conners
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jennifer R Geske
- Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Kathleen R Brandt
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Amy C Degnim
- Department of Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Celine M Vachon
- Department of Health Sciences Research, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Michael K O'Connor
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Deborah J Rhodes
- Department of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| |
Collapse
|
49
|
Pather K, Dix-Peek T, Duarte R, Chetty N, Augustine TN. Breast cancer cell-induced platelet activation is compounded by tamoxifen and anastrozole in vitro. Thromb Res 2019; 177:51-58. [PMID: 30851629 DOI: 10.1016/j.thromres.2019.02.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 02/01/2019] [Accepted: 02/22/2019] [Indexed: 01/06/2023]
Abstract
Platelet-tumour cell interaction is implicated in the initiation of breast cancer-associated thrombosis, with hormone-therapy (Tamoxifen/Anastrozole), increasing this risk. However, recent in vitro research indicates that Tamoxifen inhibits platelet activation, while the effects of Anastrozole on platelet activation are not well characterised. This study investigated platelet activation caused by Tamoxifen or Anastrozole-treated breast cancer cells in vitro. MCF7 and T47D cells were pre-treated with Tamoxifen or Anastrozole to mimic the effects of the drugs in vivo, and co-cultured with whole blood. Platelet activation was determined using flow cytometry. Platelet (CD41a+CD62P+) was determined using an interval gating strategy. Platelet morphology was visualised using scanning electron microscopy. Our results support clinical findings, showing that hormone-therapy is associated with platelet activation. Tamoxifen-treated MCF7 cells increased P-selectin expression, with ultrastructural analysis showing fully spread platelets. Conversely, Tamoxifen-treated T47D cells decreased P-selectin expression with platelets showing signs of early aggregation. Anastrozole pre-treatment decreased P-selectin expression, with treated MCF7 cells inducing platelet membrane folds and lamellipodia extension, and treated T47D cells inducing platelet aggregation and fibrin network formation indicating hypercoagulation. The findings support clinical studies. Hormone-therapy augments tumour cell-induced platelet activation, which may be linked to cell phenotype. This may have clinical implications for treatment strategies.
Collapse
Affiliation(s)
- K Pather
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193 Johannesburg, South Africa.
| | - T Dix-Peek
- Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193 Johannesburg, South Africa
| | - R Duarte
- Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193 Johannesburg, South Africa
| | - N Chetty
- Department of Molecular Medicine and Haematology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, National Health Laboratory Services (NHLS), 7 York Road, Parktown, 2193 Johannesburg, South Africa
| | - T N Augustine
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193 Johannesburg, South Africa.
| |
Collapse
|
50
|
Porsch M, Özdemir E, Wisniewski M, Graf S, Bull F, Hoffmann K, Ignatov A, Haybaeck J, Grosse I, Kalinski T, Nass N. Time resolved gene expression analysis during tamoxifen adaption of MCF-7 cells identifies long non-coding RNAs with prognostic impact. RNA Biol 2019; 16:661-674. [PMID: 30760083 DOI: 10.1080/15476286.2019.1581597] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Acquired tamoxifen resistance is a persistent problem for the treatment of estrogen receptor positive, premenopausal breast cancer patients and predictive biomarkers are still elusive. We here analyzed gene expression changes in a cellular model to identify early and late changes upon tamoxifen exposure and thereby novel prognostic biomarkers. Estrogen receptor positive MCF-7 cells were incubated with 4OH-tamoxifen (10 nM) and gene expression analyzed by array hybridization during 12 weeks. Array results were confirmed by nCounter- and qRT-PCR technique. Pathway enrichment analysis revealed that early responses concerned mainly amine synthesis and NRF2-related signaling and evolved into a stable gene expression pattern within 4 weeks characterized by changes in glucuronidation-, estrogen metabolism-, nuclear receptor- and interferon signaling pathways. As a large number of long non coding RNAs was subject to regulation, we investigated 5 of these (linc01213, linc00632 linc0992, LOC101929547 and XR_133213) in more detail. From these, only linc01213 was upregulated but all were less abundant in estrogen-receptor negative cell lines (MDA-MB 231, SKBR-3 and UACC3199). In a web-based survival analysis linc01213 and linc00632 turned out to have prognostic impact. Linc01213 was investigated further by plasmid-mediated over-expression as well as siRNA down-regulation in MCF-7 cells. Nevertheless, this had no effect on proliferation or expression of tamoxifen regulated genes, but migration was increased. In conclusion, the cellular model identified a set of lincRNAs with prognostic relevance for breast cancer. One of these, linc01213 although regulated by 4OH-tamoxifen, is not a central regulator of tamoxifen adaption, but interferes with the regulation of migration.
Collapse
Affiliation(s)
- Martin Porsch
- a Insitute of Computer Science , Martin Luther University Halle-Wittenberg , Halle , Germany.,b Institute of Human Genetics , Martin Luther University Halle-Wittenberg , Halle , Germany.,c German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig , Leipzig , Germany
| | - Esra Özdemir
- d Institute of Pathology, Otto von Guericke University Magdeburg , Magdeburg , Germany
| | - Martin Wisniewski
- d Institute of Pathology, Otto von Guericke University Magdeburg , Magdeburg , Germany
| | - Sebastian Graf
- a Insitute of Computer Science , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Fabian Bull
- a Insitute of Computer Science , Martin Luther University Halle-Wittenberg , Halle , Germany.,b Institute of Human Genetics , Martin Luther University Halle-Wittenberg , Halle , Germany.,c German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig , Leipzig , Germany
| | - Katrin Hoffmann
- b Institute of Human Genetics , Martin Luther University Halle-Wittenberg , Halle , Germany
| | - Atanas Ignatov
- e Department of Obstetrics and Gynecology , Otto von Guericke University Magdeburg , Magdeburg , Germany
| | - Johannes Haybaeck
- d Institute of Pathology, Otto von Guericke University Magdeburg , Magdeburg , Germany.,f Diagnostic and Research Institute of Pathology , Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz , Graz , Austria.,g Department of Pathology , Medical University of Innsbruck , Innsbruck , Austria
| | - Ivo Grosse
- a Insitute of Computer Science , Martin Luther University Halle-Wittenberg , Halle , Germany.,c German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig , Leipzig , Germany
| | - Thomas Kalinski
- d Institute of Pathology, Otto von Guericke University Magdeburg , Magdeburg , Germany
| | - Norbert Nass
- d Institute of Pathology, Otto von Guericke University Magdeburg , Magdeburg , Germany
| |
Collapse
|