1
|
Palma GBH, Kaur M. miRNA-128 and miRNA-223 regulate cholesterol-mediated drug resistance in breast cancer. IUBMB Life 2023; 75:743-764. [PMID: 37070323 DOI: 10.1002/iub.2726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 03/24/2023] [Indexed: 04/19/2023]
Abstract
BACKGROUND Breast cancer is the second most common malignancy worldwide and 70% of all breast cancer cases are estrogen receptor-positive (ER+). Endocrine therapy, Tamoxifen (TAM), is a popular treatment for ER+ breast cancer patients; however, despite its success in reducing breast cancer mortality, cancer drug resistance remains a significant challenge. A major contributor to this resistance is the dysregulation of cholesterol homeostasis, where breast cancer cells have elevated cholesterol levels. MicroRNAs (miRNAs) are master regulators of cholesterol-related and cancer drug resistance pathways, and their aberrant expression often confers resistance. Therefore, we aimed to investigate the roles of miRNA-128 and miRNA-223 in cholesterol-mediated TAM resistance. METHODS Three breast cancer cell lines were treated with a combination of 1 μM TAM and 10 μM of a cholesterol depleting agent (Acetyl Plumbagin: AP) following transfection with a miR-128 inhibitor or a miR-223 mimic. Cell viability and cholesterol levels were assessed using an MTT assay and fluorescence staining, respectively. In addition, expression levels of several genes and proteins involved in cancer drug resistance and cholesterol homeostasis were also assessed using RT-qPCR and western blotting. RESULTS The combination treatment with altered miRNA expression led to reduced cell viability due to a reduction in free cholesterol and lipid rafts in MCF-7, MDA-MB-231, and long-term estrogen-deprived cells (resistant breast cancer cells). Moreover, reduced miR-128 expression was favoured in all breast cancer cell lines as this alteration lowered the expression of genes involved in cholesterol synthesis and transport, drug resistance, and cell signalling. CONCLUSIONS Investigating the gene expression profiles in different breast cancer cell lines was important to elucidate further the molecular mechanisms involved in miRNA-regulated cholesterol homeostasis and cancer drug resistance. Therefore, our findings demonstrated that miR-128 and miR-223 could be potential targets in reducing TAM resistance through the depletion of excess cholesterol.
Collapse
Affiliation(s)
| | - Mandeep Kaur
- School of Molecular and Cell Biology, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
2
|
Arnachellen D, Xulu KR, Pillay K, Augustine TN. Breast Tumor Cells Evade the Cytotoxic Action of Anastrozole, Aspirin, and Clopidogrel Cocktail. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2023; 29:1205-1219. [PMID: 37749673 DOI: 10.1093/micmic/ozad032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 02/07/2023] [Accepted: 02/22/2023] [Indexed: 09/27/2023]
Abstract
Globally, breast cancer is among the most frequently diagnosed and common cause of death among women. Aromatase inhibitors, such as anastrozole, are one of the first-line therapies used in the treatment of breast cancer in postmenopausal women; however, thromboembolic complications are common. Thus, this study investigated the combined effects of anastrozole and antiplatelet therapies, aspirin and clopidogrel, on breast cancer cytotoxicity and survival in vitro. Breast cancer cell lines (MCF-7 and T47D) were treated with varying Cmax concentrations of anastrozole and/or antiplatelet therapies for 24 h. A wound-healing scratch assay was used to measure migration and the WST-1 assay for cellular proliferation. An autophagy/cytotoxicity dual staining kit was used to assay cell death and survival. Changes in cell morphology were assessed using scanning electron microscopy. Data were analyzed with Statistica software. Our findings showed that sub-phenotypic differences exist between the luminal-A breast cancer cell lines, with T47D cells being more aggressive than MCF-7 cells. Cellular proliferation and migration responded in a dose-dependent manner for the different treatment groups. Notably, anastrozole combined with aspirin and clopidogrel mediated higher levels of cell survival than each agent individually, with autophagy levels being significantly increased in comparison to that induced with antiplatelet therapy alone.
Collapse
Affiliation(s)
- Derushka Arnachellen
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193, Johannesburg, South Africa
| | - Kutlwano R Xulu
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193, Johannesburg, South Africa
| | - Kiveshen Pillay
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193, Johannesburg, South Africa
| | - Tanya N Augustine
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193, Johannesburg, South Africa
| |
Collapse
|
3
|
Banjare L, Singh Y, Verma SK, Singh AK, Kumar P, Kumar S, Jain AK, Thareja S. Multifaceted 3D-QSAR analysis for the identification of pharmacophoric features of biphenyl analogues as aromatase inhibitors. J Biomol Struct Dyn 2023; 41:1322-1341. [PMID: 34963408 DOI: 10.1080/07391102.2021.2019122] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Aromatase, a cytochrome P450 enzyme, is responsible for the conversion of androgens to estrogens, which fuel the multiplication of cancerous cells. Inhibition of estrogen biosynthesis by aromatase inhibitors (AIs) is one of the highly advanced therapeutic approach available for the treatment of estrogen-positive breast cancer. Biphenyl moiety aids lipophilicity to the conjugated scaffold and enhances the accessibility of the ligand to the target. The present study is focused on the investigation of, the mode of binding of biphenyl with aromatase, prediction of ligand-target binding affinities, and pharmacophoric features essential for favorable for aromatase inhibition. A multifaceted 3D-QSAR (SOMFA, Field and Gaussian) along with molecular docking, molecular dynamic simulations and pharmacophore mapping were performed on a series of biphenyl bearing molecules (1-33) with a wide range of aromatase inhibitory activity (0.15-920 nM). Among the generated 3D-QSAR models, the Force field-based 3D-QSAR model (R2 = 0.9151) was best as compared to SOMFA and Gaussian Field (R2=0.7706, 0.9074, respectively). However, all the generated 3D-QSAR models were statistically fit, robust enough, and reliable to explain the variation in biological activity in relation to pharmacophoric features of dataset molecules. A four-point pharmacophoric features with three acceptor sites (A), one aromatic ring (R) features, AAAR_1, were obtained with the site and survival score values 0.890 and 4.613, respectively. The generated 3D-QSAR plots in the study insight into the structure-activity relationship of dataset molecules, which may help in the designing of potent biphenyl derivatives as newer inhibitors of aromatase.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Laxmi Banjare
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur, Chhattisgarh, India
| | - Yogesh Singh
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences Central, University of Punjab, Bathinda, Punjab, India
| | - Sant Kumar Verma
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Atul Kumar Singh
- Molecular Signaling and Drug Discovery Laboratory, Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences Central, University of Punjab, Bathinda, Punjab, India
| | - Shashank Kumar
- Molecular Signaling and Drug Discovery Laboratory, Department of Biochemistry, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Akhlesh Kumar Jain
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur, Chhattisgarh, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences Central, University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
4
|
Song WM, Chia PL, Zhou X, Walsh M, Silva J, Zhang B. Pseudo-temporal dynamics of chemoresistant triple negative breast cancer cells reveal EGFR/HER2 inhibition as synthetic lethal during mid-neoadjuvant chemotherapy. iScience 2023; 26:106064. [PMID: 36824282 PMCID: PMC9942122 DOI: 10.1016/j.isci.2023.106064] [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: 09/01/2022] [Revised: 10/17/2022] [Accepted: 01/23/2023] [Indexed: 01/29/2023] Open
Abstract
In the absence of targetable hormonal axes, chemoresistance for triple-negative breast cancer (TNBC) often compromises patient outcomes. To investigate the underlying tumor dynamics, we performed trajectory analysis on the single-nuclei RNA-seq (snRNA-seq) of chemoresistant tumor clones during neoadjuvant chemotherapy (NAC). It revealed a common tumor trajectory across multiple patients with HER2-like expansions during NAC. Genome-wide CRISPR-Cas9 knock-out on mammary epithelial cells revealed chemosensitivity-promoting knock-outs were up-regulated along the tumor trajectory. Furthermore, we derived a consensus gene signature of TNBC chemoresistance by comparing the trajectory transcriptome with chemoresistant transcriptomes from TNBC cell lines and poor prognosis patient samples to predict FDA-approved drugs, including afatinib (pan-HER inhibitor), targeting the consensus signature. We validated the synergistic efficacy of afatinib and paclitaxel in chemoresistant TNBC cells and confirmed pharmacological suppression of the consensus signature. The study provides a dynamic model of chemoresistant tumor transcriptome, and computational framework for pharmacological intervention.
Collapse
Affiliation(s)
- Won-Min Song
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Pei-Ling Chia
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Martin Walsh
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Jose Silva
- Department of Pathology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
5
|
Proteomic time course of breast cancer cells highlights enhanced sensitivity to Stat3 and Src inhibitors prior to endocrine resistance development. Cancer Gene Ther 2023; 30:324-334. [PMID: 36266450 PMCID: PMC9935392 DOI: 10.1038/s41417-022-00548-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 09/28/2022] [Accepted: 10/06/2022] [Indexed: 11/08/2022]
Abstract
To prevent the development of endocrine-resistant breast cancer, additional targeted therapies are increasingly being trialled in combination with endocrine therapy. The molecular mechanisms facilitating cancer cell survival during endocrine treatment remain unknown but could help direct selection of additional targeted therapies. We present a novel proteomic timecourse dataset, profiling potential drug targets in a population of MCF7 cells during 1 year of tamoxifen treatment. Reverse phase protein arrays profiled >70 proteins across 30 timepoints. A biphasic response to tamoxifen was evident, which coincided with changes in growth rate. Tamoxifen strongly impeded cell growth for the first 160 days, followed by gradual growth recovery and eventual resistance development. The growth-impeded phase was distinguished by the phosphorylation of Stat3 (y705) and Src (y527). Tumour tissue from patients treated with neo-adjuvant endocrine therapy (<4 months) also displayed increased Stat3 and Src signalling. Inhibitors of Stat3 (napabucasin) and Src (dasatinib), were effective at killing tamoxifen-treated MCF7 and T47D cells. Sensitivity to both drugs was significantly enhanced once tamoxifen had induced the growth-impeded phase. This novel proteomic resource identifies key mechanisms enabling cell survival during tamoxifen treatment. It provides valuable insight into potential drug combinations and timing that may prevent the development of endocrine resistance.
Collapse
|
6
|
Filip-Psurska B, Zachary H, Strzykalska A, Wietrzyk J. Vitamin D, Th17 Lymphocytes, and Breast Cancer. Cancers (Basel) 2022; 14:cancers14153649. [PMID: 35954312 PMCID: PMC9367508 DOI: 10.3390/cancers14153649] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary The effect of vitamin D3 on the development of breast cancer (favorable, ineffective, or even unfavorable) depends on many factors, such as age, menopausal status, or obesity. The immunomodulatory effect of vitamin D may be unfavorable in case of breast cancer progression. The effect of vitamin D on Th17 cells may depend on disease type and patients’ age. Our goal was to summarize the data available and to find indications of vitamin D treatment failure or success. Therefore, in this review, we present data describing the effects of vitamin D3 on Th17 cells, mainly in breast cancer. Abstract Vitamin D3, which is well known to maintain calcium homeostasis, plays an important role in various cellular processes. It regulates the proliferation and differentiation of several normal cells, including immune and neoplastic cells, influences the cell cycle, and stimulates cell maturation and apoptosis through a mechanism dependent on the vitamin D receptor. The involvement of vitamin D3 in breast cancer development has been observed in numerous clinical studies. However, not all studies support the protective effect of vitamin D3 against the development of this condition. Furthermore, animal studies have revealed that calcitriol or its analogs may stimulate tumor growth or metastasis in some breast cancer models. It has been postulated that the effect of vitamin D3 on T helper (Th) 17 lymphocytes is one of the mechanisms promoting metastasis in these murine models. Herein we present a literature review on the existing data according to the interplay between vitamin D, Th17 cell and breast cancer. We also discuss the effects of this vitamin on Th17 lymphocytes in various disease entities known to date, due to the scarcity of scientific data on Th17 lymphocytes and breast cancer. The presented data indicate that the effect of vitamin D3 on breast cancer development depends on many factors, such as age, menopausal status, or obesity. According to that, more extensive clinical trials and studies are needed to assess the importance of vitamin D in breast cancer, especially when no correlations seem to be obvious.
Collapse
|
7
|
Cheng GJ, Leung EY, Singleton DC. In vitro breast cancer models for studying mechanisms of resistance to endocrine therapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:297-320. [PMID: 36045910 PMCID: PMC9400723 DOI: 10.37349/etat.2022.00084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/24/2022] [Indexed: 11/19/2022] Open
Abstract
The development of endocrine resistance is a common reason for the failure of endocrine therapies in hormone receptor-positive breast cancer. This review provides an overview of the different types of in vitro models that have been developed as tools for studying endocrine resistance. In vitro models include cell lines that have been rendered endocrine-resistant by ex vivo treatment; cell lines with de novo resistance mechanisms, including genetic alterations; three-dimensional (3D) spheroid, co-culture, and mammosphere techniques; and patient-derived organoid models. In each case, the key discoveries, different analysis strategies that are suitable, and strengths and weaknesses are discussed. Certain recently developed methodologies that can be used to further characterize the biological changes involved in endocrine resistance are then emphasized, along with a commentary on the types of research outcomes that using these techniques can support. Finally, a discussion anticipates how these recent developments will shape future trends in the field. We hope this overview will serve as a useful resource for investigators that are interested in understanding and testing hypotheses related to mechanisms of endocrine therapy resistance.
Collapse
Affiliation(s)
- Gary J. Cheng
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand
| | - Euphemia Y. Leung
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland 1023, New Zealand
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland 1023, New Zealand
| | - Dean C. Singleton
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1023, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland 1023, New Zealand
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland 1023, New Zealand
| |
Collapse
|
8
|
Shah V, Bhaliya J, Patel GM. In silico docking and ADME study of deketene curcumin derivatives (DKC) as an aromatase inhibitor or antagonist to the estrogen-alpha positive receptor (Erα+): potent application of breast cancer. Struct Chem 2022; 33:571-600. [PMID: 35106036 PMCID: PMC8794617 DOI: 10.1007/s11224-021-01871-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/13/2021] [Indexed: 11/26/2022]
Abstract
Regardless of many extensive studies, hormonal-based breast cancer is the most common cause of cancer-related mortality of females worldwide. Indeed, estrogen receptor-positive (ER +) is the communal subtype in breast cancer. To treat this, three types of medications are typically used: selective estrogen receptor modulators (SERMs), selective estrogen receptor down modulators (SERDMs), and aromatase inhibitors (AIs), all of which directly interact with the activation of the estrogen signaling pathway and its formation. Despite their effectiveness, the development of new treatments is required since clinical efficacy is restricted owing to resistance. As a result, in silico studies for drug discovery are booming over the decades because of their affordability and less time-consuming features. Here, 25 deketene curcumin derivatives have been selected for docking studies through MVD software over the positive type of breast cancer through both the treatment hosts Erα + receptor and aromatase. DKC compounds are used because they have several pharmacological uses, including anti-cancer, anti-diabetic, anti-viral, anti-fungal, and anti-bacterial properties. Moreover, an ADME study was carried out for DKC derivatives that reveal the optimum drug-likeness profile. From 25 derivatives, the results showed a better MolDock score, hydrogen bonding, and steric interaction between compounds DKC-10, DKC-20, and DKC-21 with Erα + and aromatase. Although the study was done on both the treatable path hosts, better results were obtained with Erα + as an antagonist. Therefore, it is proposed that three selected DKC derivatives would be better therapeutic agents against breast cancer.
Collapse
Affiliation(s)
- Vraj Shah
- Department of Chemistry, School of Science, ITM SLS Baroda University, Vadodara, 391510 India
| | - Jaydip Bhaliya
- Department of Chemistry, School of Science, ITM SLS Baroda University, Vadodara, 391510 India
| | - Gautam M. Patel
- Department of Industrial Chemistry, Institute of Science & Technology for Advanced Studies & Research (ISTAR), CVM University, V.V., Nagar, 388120 GJ India
| |
Collapse
|
9
|
Lewoniewska S, Oscilowska I, Huynh TYL, Prokop I, Baszanowska W, Bielawska K, Palka J. Troglitazone-Induced PRODH/POX-Dependent Apoptosis Occurs in the Absence of Estradiol or ERβ in ER-Negative Breast Cancer Cells. J Clin Med 2021; 10:jcm10204641. [PMID: 34682765 PMCID: PMC8538344 DOI: 10.3390/jcm10204641] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/28/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary PRODH/POX (proline dehydrogenase/proline oxidase) is a mitochondrial enzyme that catalyzes proline degradation generating reactive oxygen species (ROS). Estrogens limit proline availability for PRODH/POX by stimulating collagen biosynthesis. It has been considered that estrogens determine efficiency of troglitazone (TGZ)-induced PRODH/POX-dependent apoptosis in breast cancer cells. The studies were performed in wild-type and PRODH/POX-silenced estrogen-dependent MCF-7 cells and estrogen-independent MDA-MB-231 cells. DNA and collagen biosynthesis were determined by radiometric method, ROS production was measured by fluorescence assay, protein expression was determined by Western blot and proline concentration by LC/MS analysis. We found that: i/TGZ-induced apoptosis in MDA-MB-231 occurs only in the absence of estradiol or ERβ, ii/the process is mediated by PRODH/POX, iii/and is facilitated by proline availability for PRODH/POX by TGZ-dependent inhibition of collagen biosynthesis (proline utilizing process). The data suggest that combined TGZ and anti-estrogen treatment could be considered in experimental therapy of ER negative breast cancers. Abstract The impact of estradiol on troglitazone (TGZ)-induced proline dehydrogenase/proline oxidase (PRODH/POX)-dependent apoptosis was studied in wild-type and PRODH/POX-silenced estrogen receptor (ER) dependent MCF-7 cells and ER-independent MDA-MB-231 cells. DNA and collagen biosynthesis were determined by radiometric method, prolidase activity evaluated by colorimetric method, ROS production was measured by fluorescence assay. Protein expression was determined by Western blot and proline concentration by LC/MS analysis. PRODH/POX degrades proline yielding reactive oxygen species (ROS). Estrogens stimulate collagen biosynthesis utilizing free proline and limiting its availability for PRODH/POX-dependent apoptosis. TGZ cytotoxicity was highly pronounced in wild-type MDA-MB-231 cells cultured in medium without estradiol or in the cells cultured in medium with estradiol but deprived of ERβ (by ICI-dependent degradation), while in PRODH/POX-silenced cells the process was not affected. The TGZ cytotoxicity was accompanied by increase in PRODH/POX expression, ROS production, expression of cleaved caspase-3, caspase-9 and PARP, inhibition of collagen biosynthesis, prolidase activity and decrease in intracellular proline concentration. The phenomena were not observed in PRODH/POX-silenced cells. The data suggest that TGZ-induced apoptosis in MDA-MB-231 cells cultured in medium without estradiol or deprived of ERβ is mediated by PRODH/POX and the process is facilitated by proline availability for PRODH/POX by TGZ-dependent inhibition of collagen biosynthesis. It suggests that combined TGZ and antiestrogen treatment could be considered in experimental therapy of estrogen receptor negative breast cancers.
Collapse
Affiliation(s)
- Sylwia Lewoniewska
- Department of Medicinal Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland; (S.L.); (T.Y.L.H.); (I.P.); (W.B.); (K.B.)
| | - Ilona Oscilowska
- Department of Analysis and Bioanalysis of Medicines, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland;
| | - Thi Yen Ly Huynh
- Department of Medicinal Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland; (S.L.); (T.Y.L.H.); (I.P.); (W.B.); (K.B.)
| | - Izabela Prokop
- Department of Medicinal Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland; (S.L.); (T.Y.L.H.); (I.P.); (W.B.); (K.B.)
| | - Weronika Baszanowska
- Department of Medicinal Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland; (S.L.); (T.Y.L.H.); (I.P.); (W.B.); (K.B.)
| | - Katarzyna Bielawska
- Department of Medicinal Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland; (S.L.); (T.Y.L.H.); (I.P.); (W.B.); (K.B.)
| | - Jerzy Palka
- Department of Medicinal Chemistry, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland; (S.L.); (T.Y.L.H.); (I.P.); (W.B.); (K.B.)
- Correspondence: ; Tel.: +48-85-748-5706
| |
Collapse
|
10
|
Clarke R, Jones BC, Sevigny CM, Hilakivi-Clarke LA, Sengupta S. Experimental models of endocrine responsive breast cancer: strengths, limitations, and use. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:762-783. [PMID: 34532657 PMCID: PMC8442978 DOI: 10.20517/cdr.2021.33] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Breast cancers characterized by expression of estrogen receptor-alpha (ER; ESR1) represent approximately 70% of all new cases and comprise the largest molecular subtype of this disease. Despite this high prevalence, the number of adequate experimental models of ER+ breast cancer is relatively limited. Nonetheless, these models have proved very useful in advancing understanding of how cells respond to and resist endocrine therapies, and how the ER acts as a transcription factor to regulate cell fate signaling. We discuss the primary experimental models of ER+ breast cancer including 2D and 3D cultures of established cell lines, cell line- and patient-derived xenografts, and chemically induced rodent models, with a consideration of their respective general strengths and limitations. What can and cannot be learned easily from these models is also discussed, and some observations on how these models may be used more effectively are provided. Overall, despite their limitations, the panel of models currently available has enabled major advances in the field, and these models remain central to the ability to study mechanisms of therapy action and resistance and for hypothesis testing that would otherwise be intractable or unethical in human subjects.
Collapse
Affiliation(s)
- Robert Clarke
- The Hormel Institute and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Austin, MN 55912, USA
| | - Brandon C Jones
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Catherine M Sevigny
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA.,The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Leena A Hilakivi-Clarke
- The Hormel Institute and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Austin, MN 55912, USA
| | - Surojeet Sengupta
- The Hormel Institute and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Austin, MN 55912, USA
| |
Collapse
|
11
|
Banjare L, Verma SK, Jain AK, Thareja S. Design and pharmacophoric identification of flavonoid scaffold‐based aromatase inhibitors. J Heterocycl Chem 2020. [DOI: 10.1002/jhet.4068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Laxmi Banjare
- School of Pharmaceutical SciencesGuru Ghasidas Central University Bilaspur Chhattisgarh India
| | - Sant Kumar Verma
- School of Pharmaceutical SciencesGuru Ghasidas Central University Bilaspur Chhattisgarh India
| | - Akhlesh Kumar Jain
- School of Pharmaceutical SciencesGuru Ghasidas Central University Bilaspur Chhattisgarh India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural ProductsCentral University of Punjab Bathinda Punjab India
| |
Collapse
|
12
|
Gelsomino L, Giordano C, La Camera G, Sisci D, Marsico S, Campana A, Tarallo R, Rinaldi A, Fuqua S, Leggio A, Grande F, Bonofiglio D, Andò S, Barone I, Catalano S. Leptin Signaling Contributes to Aromatase Inhibitor Resistant Breast Cancer Cell Growth and Activation of Macrophages. Biomolecules 2020; 10:biom10040543. [PMID: 32260113 PMCID: PMC7226081 DOI: 10.3390/biom10040543] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/23/2020] [Accepted: 04/01/2020] [Indexed: 12/22/2022] Open
Abstract
Obesity represents a risk factor for breast cancer development and therapy resistance, but the molecular players underling these links are unclear. Here, we identify a role for the obesity-cytokine leptin in sustaining aromatase inhibitor (AI) resistant growth and progression in breast cancer. Using as experimental models MCF-7 breast cancer cells surviving long-term treatment with the AI anastrozole (AnaR) and Ana-sensitive counterparts, we found that AnaR cells expressed higher levels of leptin and its receptors (ObR) along with a constitutive activation of downstream effectors. Accordingly, leptin signaling inhibition reduced only AnaR cell growth and motility, highlighting the existence of an autocrine loop in mechanisms governing drug-resistant phenotypes. In agreement with ObR overexpression, increasing doses of leptin were able to stimulate to a greater extent growth and migration in AnaR than sensitive cells. Moreover, leptin contributed to enhanced crosstalk between AnaR cells and macrophages within the tumor microenvironment. Indeed, AnaR, through leptin secretion, modulated macrophage profiles and increased macrophage motility through CXCR4 signaling, as evidenced by RNA-sequencing, real-time PCR, and immunoblotting. Reciprocally, activated macrophages increased AnaR cell growth and motility in coculture systems. In conclusion, acquired AI resistance is accompanied by the development of a leptin-driven phenotype, highlighting the potential clinical benefit of targeting this cytokine network in hormone-resistant breast cancers, especially in obese women.
Collapse
Affiliation(s)
- Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (L.G.); (C.G.); (G.L.C.); (D.S.); (S.M.); (A.C.); (A.L.); (F.G.); (D.B.); (S.A.)
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (L.G.); (C.G.); (G.L.C.); (D.S.); (S.M.); (A.C.); (A.L.); (F.G.); (D.B.); (S.A.)
| | - Giusi La Camera
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (L.G.); (C.G.); (G.L.C.); (D.S.); (S.M.); (A.C.); (A.L.); (F.G.); (D.B.); (S.A.)
| | - Diego Sisci
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (L.G.); (C.G.); (G.L.C.); (D.S.); (S.M.); (A.C.); (A.L.); (F.G.); (D.B.); (S.A.)
| | - Stefania Marsico
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (L.G.); (C.G.); (G.L.C.); (D.S.); (S.M.); (A.C.); (A.L.); (F.G.); (D.B.); (S.A.)
| | - Antonella Campana
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (L.G.); (C.G.); (G.L.C.); (D.S.); (S.M.); (A.C.); (A.L.); (F.G.); (D.B.); (S.A.)
| | - Roberta Tarallo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi (SA), Italy; (R.T.); (A.R.)
| | - Antonio Rinaldi
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi (SA), Italy; (R.T.); (A.R.)
| | - Suzanne Fuqua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, MS: 600 N1220.01 Alkek Building, Houston, TX 77030, USA;
| | - Antonella Leggio
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (L.G.); (C.G.); (G.L.C.); (D.S.); (S.M.); (A.C.); (A.L.); (F.G.); (D.B.); (S.A.)
| | - Fedora Grande
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (L.G.); (C.G.); (G.L.C.); (D.S.); (S.M.); (A.C.); (A.L.); (F.G.); (D.B.); (S.A.)
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (L.G.); (C.G.); (G.L.C.); (D.S.); (S.M.); (A.C.); (A.L.); (F.G.); (D.B.); (S.A.)
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (L.G.); (C.G.); (G.L.C.); (D.S.); (S.M.); (A.C.); (A.L.); (F.G.); (D.B.); (S.A.)
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (L.G.); (C.G.); (G.L.C.); (D.S.); (S.M.); (A.C.); (A.L.); (F.G.); (D.B.); (S.A.)
- Correspondence: (I.B.); (S.C.); Tel.: +39-0984-496216 (I.B.); +39-0984-496207 (S.C.)
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (L.G.); (C.G.); (G.L.C.); (D.S.); (S.M.); (A.C.); (A.L.); (F.G.); (D.B.); (S.A.)
- Correspondence: (I.B.); (S.C.); Tel.: +39-0984-496216 (I.B.); +39-0984-496207 (S.C.)
| |
Collapse
|
13
|
Creevey L, Bleach R, Madden SF, Toomey S, Bane FT, Varešlija D, Hill AD, Young LS, McIlroy M. Altered Steroid Milieu in AI-Resistant Breast Cancer Facilitates AR Mediated Gene-Expression Associated with Poor Response to Therapy. Mol Cancer Ther 2019; 18:1731-1743. [PMID: 31289138 DOI: 10.1158/1535-7163.mct-18-0791] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/23/2018] [Accepted: 07/03/2019] [Indexed: 11/16/2022]
Abstract
Divergent roles for androgen receptor (AR) in breast cancer have been reported. Following aromatase inhibitor (AI) treatment, the conversion of circulating androgens into estrogens can be diminished by >99%. We wished to establish whether the steroid environment can dictate the role of AR and the implications of this for subsequent therapy. This study utilizes models of AI resistance to explore responsiveness to PI3K/mTOR and anti-AR therapy when cells are exposed to unconverted weak androgens. Transcriptomic alterations driven by androstenedione (4AD) were assessed by RNA-sequencing. AR and estrogen receptor (ER) recruitment to target gene promoters was evaluated using ChIP, and relevance to patient profiles was performed using publicly available data sets. Although BEZ235 showed decreased viability across AI-sensitive and -resistant cell lines, anti-AR treatment elicited a decrease in cell viability only in the AI-resistant model. Serum and glucocorticoid-regulated kinase 3 (SGK3) and cAMP-dependent protein kinase inhibitor β (PKIB) were confirmed to be regulated by 4AD and shown to be mediated by AR; crucially, reexposure to estradiol suppressed expression of these genes. Meta-analysis of transcript levels showed high expression of SGK3 and PKIB to be associated with poor response to endocrine therapy (HR = 2.551, P = 0.003). Furthermore, this study found levels of SGK3 to be sustained in patients who do not respond to AI therapy. This study highlights the importance of the tumor steroid environment. SGK3 and PKIB are associated with poor response to endocrine therapy and could have utility in tailoring therapeutic approaches.
Collapse
Affiliation(s)
- Laura Creevey
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Rachel Bleach
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Stephen F Madden
- Data Science Centre, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Sinead Toomey
- Department of Oncology, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Fiona T Bane
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Damir Varešlija
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Arnold D Hill
- Department of Surgery, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Leonie S Young
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Marie McIlroy
- Endocrine Oncology Research Group, Department of Surgery, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland.
| |
Collapse
|
14
|
Banjare L, Verma SK, Jain AK, Thareja S. Structure Guided Molecular Docking Assisted Alignment Dependent 3DQSAR Study on Steroidal Aromatase Inhibitors (SAIs) as Anti-breast Cancer Agents. LETT DRUG DES DISCOV 2019. [DOI: 10.2174/1570180815666181010101024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background:
In spite of the availability of various treatment approaches including
surgery, radiotherapy, and hormonal therapy, the steroidal aromatase inhibitors (SAIs) play a
significant role as chemotherapeutic agents for the treatment of estrogen-dependent breast cancer
with the benefit of reduced risk of recurrence. However, due to greater toxicity and side effects
associated with currently available anti-breast cancer agents, there is emergent requirement to
develop target-specific AIs with safer anti-breast cancer profile.
Methods:
It is challenging task to design target-specific and less toxic SAIs, though the molecular
modeling tools viz. molecular docking simulations and QSAR have been continuing for more than
two decades for the fast and efficient designing of novel, selective, potent and safe molecules
against various biological targets to fight the number of dreaded diseases/disorders. In order to
design novel and selective SAIs, structure guided molecular docking assisted alignment dependent
3D-QSAR studies was performed on a data set comprises of 22 molecules bearing steroidal
scaffold with wide range of aromatase inhibitory activity.
Results:
3D-QSAR model developed using molecular weighted (MW) extent alignment approach
showed good statistical quality and predictive ability when compared to model developed using
moments of inertia (MI) alignment approach.
Conclusion:
The explored binding interactions and generated pharmacophoric features (steric and
electrostatic) of steroidal molecules could be exploited for further design, direct synthesis and
development of new potential safer SAIs, that can be effective to reduce the mortality and
morbidity associated with breast cancer.
Collapse
Affiliation(s)
- Laxmi Banjare
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495 009, C.G., India
| | - Sant Kumar Verma
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495 009, C.G., India
| | - Akhlesh Kumar Jain
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495 009, C.G., India
| | - Suresh Thareja
- School of Pharmaceutical Sciences, Guru Ghasidas Central University, Bilaspur- 495 009, C.G., India
| |
Collapse
|
15
|
Merkulov VM, Leberfarb EY, Merkulova TI. Regulatory SNPs and their widespread effects on the transcriptome. J Biosci 2018; 43:1069-1075. [PMID: 30541964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Currently, it is generally accepted that the cis-acting effects of noncoding variants on gene expression are a major factor for phenotypic variation in complex traits and disease susceptibility. Meanwhile, the protein products of many target genes for the identified cis-regulatory variants (rSNPs) are regulatory molecules themselves (transcription factors, effectors, components of signal transduction pathways, etc.), which implies dramatic downstream effects of these variations on complex gene networks. Here, we brief the results of recent most comprehensive studies on the role of rSNPs in transcriptional regulation across the genome.
Collapse
Affiliation(s)
- Vasily M Merkulov
- Laboratory of Gene Expression Regulation, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | | | | |
Collapse
|
16
|
Merkulov VM, Leberfarb EY, Merkulova TI. Regulatory SNPs and their widespread effects on the transcriptome. J Biosci 2018. [DOI: 10.1007/s12038-018-9817-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Petrossian K, Kanaya N, Lo C, Hsu PY, Nguyen D, Yang L, Yang L, Warden C, Wu X, Pillai R, Bernal L, Huang CS, Kruper L, Yuan Y, Somlo G, Mortimer J, Chen S. ERα-mediated cell cycle progression is an important requisite for CDK4/6 inhibitor response in HR+ breast cancer. Oncotarget 2018; 9:27736-27751. [PMID: 29963233 PMCID: PMC6021239 DOI: 10.18632/oncotarget.25552] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/19/2018] [Indexed: 01/01/2023] Open
Abstract
While ER has multiple biological effects, ER-cyclin D1-CDK4/6-RB is a critical pathway for the action of estrogen on the cell cycle, especially for breast cancers that rely on estrogen for growth. The latest and most efficient CDK4/6 inhibitors target the phosphorylation of retinoblastoma (RB) tumor suppressor gene; thus, altering levels of many cell cycle molecules. Estrogen receptor (ER)+/HER2- breast cancers have shown great progression free survival when CDK4/6 inhibitors are combined with endocrine therapies. Here we report the mechanism of antiestrogen (fulvestrant) combination with CDK4/6 inhibitors is due to synergism in the suppression of ER-mediated cell cycle progression. Furthermore, we performed single cell analysis of cells from an estrogen dependent/hormone receptor-positive patient derived xenograft (PDX) tumor model treated with palbociclib. These single cells expressed various levels of ER and RB which are involved in cell cycle regulation; and the response to palbociclib treatment relies not only on the ER-cyclin D1-CDK4/6-RB pathway but it is also dependent on elevated levels of ER and/or RB. Our preclinical studies show that palbociclib response is dependent on cells with ER, which is directly involved in cell cycle progression in hormone receptor positive (HR+) breast cancer.
Collapse
Affiliation(s)
- Karineh Petrossian
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Noriko Kanaya
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Chiao Lo
- Department of Breast Health, National Taiwan University Hospital, Taipei City, Taiwan
| | - Pei-Yin Hsu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Duc Nguyen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Lixin Yang
- Molecular Pathology Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Lu Yang
- Integrative Genomics Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Charles Warden
- Integrative Genomics Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Xiwei Wu
- Integrative Genomics Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Raju Pillai
- Molecular Pathology Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Lauren Bernal
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Chiun-Sheng Huang
- Department of Breast Health, National Taiwan University Hospital, Taipei City, Taiwan
| | - Laura Kruper
- Department of Surgery, City of Hope Medical Center, Duarte, CA, United States
| | - Yuan Yuan
- Department of Medical Oncology and Therapeutics Research, City of Hope Medical Center, Duarte, CA, United States
| | - George Somlo
- Department of Medical Oncology and Therapeutics Research, City of Hope Medical Center, Duarte, CA, United States
| | - Joanne Mortimer
- Department of Medical Oncology and Therapeutics Research, City of Hope Medical Center, Duarte, CA, United States
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| |
Collapse
|
18
|
Hawsawi Y, Humphries MP, Wright A, Berwick A, Shires M, Al-Kharobi H, El-Gendy R, Jove M, Twelves C, Speirs V, Beattie J. Deregulation of IGF-binding proteins -2 and -5 contributes to the development of endocrine resistant breast cancer in vitro. Oncotarget 2017; 7:32129-43. [PMID: 27050076 PMCID: PMC5078002 DOI: 10.18632/oncotarget.8534] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 03/14/2016] [Indexed: 12/28/2022] Open
Abstract
Tamoxifen (TAM) remains the adjuvant therapy of choice for pre-menopausal women with ERα-positive breast cancer. Resistance and recurrence remain, however, a major challenge with many women relapsing and subsequently dying. The insulin-like growth factor (IGF) axis is involved in breast cancer pathogenesis and progression to endocrine resistant disease, but there is very little data on the expression and potential role of IGF-binding proteins (IGFBP) during acquisition of the resistant phenotype. The aim of this study was to determine the expression and functional role of IGFBP-2 and -5 in the development of TAM resistance (TamR) in vitro and to test retrospectively whether they were predictive of resistance in a tissue microarray of 77 women with primary breast cancers who relapsed on/after endocrine therapy and 193 who did not with long term follow up. Reciprocal expression of IGFBP-2 and IGFBP-5 was observed at both mRNA and protein level in TamR cells. IGFBP-2 expression was increased by 10-fold while IGFBP-5 was decreased by 100-fold, compared to TAM-sensitive control cells. shRNA-mediated silencing of IGFBP-2 in TamR cells restored TAM sensitivity suggesting a causal role for this gene in TamR. While silencing of IGFBP-5 in control cells had no effect on TAM sensitivity, it significantly increased the migratory capacity of these cells. Quantitative image analysis of immunohistochemical data failed, however, to demonstrate an effect of IGFBP2 expression in endocrine-relapsed patients. Likewise, IGFBP-2 and IGFBP-5 expression failed to show any significant associations with survival either in patients relapsing or those not relapsing on/after endocrine therapy. By contrast, in silico mining of a separate published dataset showed that in patients who received endocrine treatment, loss of expression of IGBP-5 was significantly associated with worse survival. Overall these data suggest that co-ordinated and reciprocal alteration in IGFBP-2 and −5 expression may play a role in the acquisition of endocrine resistance.
Collapse
Affiliation(s)
- Yousef Hawsawi
- Department of Oral Biology, St James's University Hospital, Leeds, UK.,Leeds Institute of Cancer and Pathology, University of Leeds, UK.,Current address: Department of Breast Medical Oncology, MD Anderson Cancer Centre, University of Texas, Houston, USA
| | | | - Alexander Wright
- Leeds Institute of Cancer and Pathology, University of Leeds, UK
| | - Angelene Berwick
- Leeds Institute of Cancer and Pathology, University of Leeds, UK
| | - Mike Shires
- Leeds Institute of Cancer and Pathology, University of Leeds, UK
| | - Hanaa Al-Kharobi
- Department of Oral Biology, St James's University Hospital, Leeds, UK
| | - Reem El-Gendy
- Department of Oral Biology, St James's University Hospital, Leeds, UK
| | - Maria Jove
- St James's Institute of Oncology, St James's University Hospital, Leeds, UK
| | - Chris Twelves
- St James's Institute of Oncology, St James's University Hospital, Leeds, UK.,Leeds Institute of Cancer and Pathology, University of Leeds, UK
| | - Valerie Speirs
- Leeds Institute of Cancer and Pathology, University of Leeds, UK
| | - James Beattie
- Department of Oral Biology, St James's University Hospital, Leeds, UK
| |
Collapse
|
19
|
Kumavath R, Azad M, Devarapalli P, Tiwari S, Kar S, Barh D, Azevedo V, Kumar AP. Novel aromatase inhibitors selection using induced fit docking and extra precision methods: Potential clinical use in ER-alpha-positive breast cancer. Bioinformation 2016; 12:324-331. [PMID: 28293075 PMCID: PMC5320928 DOI: 10.6026/97320630012324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 05/25/2016] [Accepted: 08/12/2016] [Indexed: 11/29/2022] Open
Abstract
Aromatase (CYP19A1) the key enzyme of estrogen biosynthesis, is often deregulated in breast cancer patients. It catalyzes the conversion of androgen to estrogen, thus responsible for production of estrogen in human body. However, it causes over-production of estrogen which eventually leads to proliferation of breast cancer cells. Identification of new small molecule inhibitors targeted against CYP19A1 therefore, facilitates to increase drug sensitivity of cancer cells. In this scenario, the present study aims to identify new molecules which could block or suppress the activity of aromatase enzyme by molecular docking studies using Schrödinger-Maestro v9.3. In this study we used in silico approach by modeling CYP19A1 protein the strcture was subjected to protein preparation wizard; to add hydrogen and optimize the protonation states of Thr310 and Ser478 and Asp309 residues. Active site of the CYP19A1 protein was identified using SiteMap tool of Scchrodinger package. We further carried out docking studies by means of Glid, with various ligands. Based on glid score, potential ligands were screeened and their interaction with CYP19A1 was identified. The best hits were further screened for Lipinski's rule for drug-likeliness and bioactivity scoring properties. Thus, we report two rubivivaxin and rhodethrin compounds that have successfully satisfied all in silico parameters, necessitating further in vitro and in vivo studies.
Collapse
Affiliation(s)
- Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences,Central University of Kerala, Padannakad P.O., Kasaragod-671314, Kerala, India
| | - Manan Azad
- Department of Genomic Science, School of Biological Sciences,Central University of Kerala, Padannakad P.O., Kasaragod-671314, Kerala, India
| | - Pratap Devarapalli
- Department of Genomic Science, School of Biological Sciences,Central University of Kerala, Padannakad P.O., Kasaragod-671314, Kerala, India
| | - Sandeep Tiwari
- Instituto de CiênciasBiológicas, Universidade Federal de Minas Gerais. MG,Brazil
| | - Shreya Kar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore-117599
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri-721172, PurbaMedinipur, West Bengal, India
| | - Vasco Azevedo
- Instituto de CiênciasBiológicas, Universidade Federal de Minas Gerais. MG,Brazil
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore-117599
- National University Cancer Institute, National University Health System, Singapore-119074
- Department of Biological Sciences, University of North Texas, Denton-762035017, Texas, USA
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore-117600
- Faculty of Health Sciences, School of Biomedical Sciences, Curtin University, Bently, Western Australia-6102
| |
Collapse
|
20
|
SERPINA1 is a direct estrogen receptor target gene and a predictor of survival in breast cancer patients. Oncotarget 2016; 6:25815-27. [PMID: 26158350 PMCID: PMC4694868 DOI: 10.18632/oncotarget.4441] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 06/19/2015] [Indexed: 01/02/2023] Open
Abstract
Of all breast cancer patients, about 70% are ER+ and 10% are ER+/HER2+. The ER+/HER2+ patients have a worse outcome compared to ER+/HER2- patients. Currently there is a lack of effective prognosis biomarkers for the prediction of outcome in ER+/HER2+ patients. Genome-wide differences in ER binding between the endocrine-responsive and endocrine-resistant cells were discovered using ChIP-seq, and combined with gene expression microarray data to identify direct ER target genes. These genes were correlated to survival outcome using publicly available breast cancer patient cohorts. We found the expression of the gene SERPINA1 to have a significant predictive value for the overall survival (OS) of ER+ patients in the TCGA cohort, and validated this finding in the Curtis cohort. SERPINA1 also has a significant predictive value for the OS of ER+/HER2+ patients in the TCGA cohort, with validation in the Bild cohort. The expression of SERPINA1 can be suppressed by fulvestrant and HER2 siRNA. Our results indicate that ER is constitutively activated, resulting in an E2-independent ER binding to the SERPINA1 gene and upregulation of SERPINA1 expression. Importantly, results of survival correlation suggests that high expression of SERPINA1 could be predictive for a better clinical outcome of ER+ and ER+/HER2+ patients.
Collapse
|
21
|
Chan HJ, Petrossian K, Chen S. Structural and functional characterization of aromatase, estrogen receptor, and their genes in endocrine-responsive and -resistant breast cancer cells. J Steroid Biochem Mol Biol 2016; 161:73-83. [PMID: 26277097 PMCID: PMC4752924 DOI: 10.1016/j.jsbmb.2015.07.018] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 07/22/2015] [Accepted: 07/29/2015] [Indexed: 11/22/2022]
Abstract
Aromatase and estrogen receptor α (ER) are two key proteins for the proliferation of endocrine-responsive and -resistant breast cancers. Aromatase is an enzyme involved in the conversion of androgen (such as testosterone) to estrogen (such as 17β-estradiol). It is also a very effective therapeutic target for the treatment of endocrine-responsive breast cancer. Comparing endocrine-responsive and -resistant breast cancer, aromatase protein levels do not change significantly. Aromatase activity; however, can be increased via PI3K/Akt/IGFR signaling pathways in endocrine resistant cells. The activity of aromatase has been reported to be modulated by phosphorylation. The ER is an important steroid nuclear receptor in the proliferation of both endocrine-responsive and -resistant cells. Although the mutation or amplification of ER can cause endocrine resistance, it is not commonly found. Some point mutations and translocation events have been characterized and shown to promote estrogen-independent growth. Phosphorylation by cross-talk with growth factor pathways is one of the main mechanisms for ligand-independent activation of ER. Taken together, both ER and aromatase are important in ER-dependent breast cancer and the development of endocrine resistance.
Collapse
Affiliation(s)
- Hei Jason Chan
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Karineh Petrossian
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States.
| |
Collapse
|
22
|
Krisnamurti DGB, Louisa M, Anggraeni E, Wanandi SI. Drug Efflux Transporters Are Overexpressed in Short-Term Tamoxifen-Induced MCF7 Breast Cancer Cells. Adv Pharmacol Sci 2016; 2016:6702424. [PMID: 26981116 PMCID: PMC4769734 DOI: 10.1155/2016/6702424] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/11/2016] [Accepted: 01/13/2016] [Indexed: 11/17/2022] Open
Abstract
Tamoxifen is the first line drug used in the treatment of estrogen receptor-positive (ER+) breast cancer. The development of multidrug resistance (MDR) to tamoxifen remains a major challenge in the treatment of cancer. One of the mechanisms related to MDR is decrease of drug influx via overexpression of drug efflux transporters such as P-glycoprotein (P-gp/MDR1), multidrug resistance associated protein (MRP), or BCRP (breast cancer resistance protein). We aimed to investigate whether the sensitivity of tamoxifen to the cells is maintained through the short period and whether the expressions of several drug efflux transporters have been upregulated. We exposed MCF7 breast cancer cells with tamoxifen 1 μM for 10 passages (MCF7 (T)). The result showed that MCF7 began to lose their sensitivity to tamoxifen from the second passage. MCF7 (T) also showed a significant increase in all transporters examined compared with MCF7 parent cells. The result also showed a significant increase of CC50 in MCF7 (T) compared to that in MCF7 (97.54 μM and 3.04 μM, resp.). In conclusion, we suggest that the expression of several drug efflux transporters such as P-glycoprotein, MRP2, and BCRP might be used and further studied as a marker in the development of tamoxifen resistance.
Collapse
Affiliation(s)
| | - Melva Louisa
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Indonesia, Jakarta 10430, Indonesia
| | - Erlia Anggraeni
- Master Program in Biomedicine, Faculty of Medicine, University of Indonesia, Jakarta 10430, Indonesia
| | - Septelia Inawati Wanandi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Indonesia, Jakarta 10430, Indonesia
| |
Collapse
|
23
|
Huang R, Xia M, Sakamuru S, Zhao J, Shahane SA, Attene-Ramos M, Zhao T, Austin CP, Simeonov A. Modelling the Tox21 10 K chemical profiles for in vivo toxicity prediction and mechanism characterization. Nat Commun 2016; 7:10425. [PMID: 26811972 PMCID: PMC4777217 DOI: 10.1038/ncomms10425] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/10/2015] [Indexed: 02/06/2023] Open
Abstract
Target-specific, mechanism-oriented in vitro assays post a promising alternative to traditional animal toxicology studies. Here we report the first comprehensive analysis of the Tox21 effort, a large-scale in vitro toxicity screening of chemicals. We test ∼ 10,000 chemicals in triplicates at 15 concentrations against a panel of nuclear receptor and stress response pathway assays, producing more than 50 million data points. Compound clustering by structure similarity and activity profile similarity across the assays reveals structure-activity relationships that are useful for the generation of mechanistic hypotheses. We apply structural information and activity data to build predictive models for 72 in vivo toxicity end points using a cluster-based approach. Models based on in vitro assay data perform better in predicting human toxicity end points than animal toxicity, while a combination of structural and activity data results in better models than using structure or activity data alone. Our results suggest that in vitro activity profiles can be applied as signatures of compound mechanism of toxicity and used in prioritization for more in-depth toxicological testing.
Collapse
Affiliation(s)
- Ruili Huang
- Division of Pre-clinical Innovation, National Center for
Advancing Translational Sciences, National Institutes of Health, 9800 Medical
Center Drive, Rockville, Maryland
20850, USA
| | - Menghang Xia
- Division of Pre-clinical Innovation, National Center for
Advancing Translational Sciences, National Institutes of Health, 9800 Medical
Center Drive, Rockville, Maryland
20850, USA
| | - Srilatha Sakamuru
- Division of Pre-clinical Innovation, National Center for
Advancing Translational Sciences, National Institutes of Health, 9800 Medical
Center Drive, Rockville, Maryland
20850, USA
| | - Jinghua Zhao
- Division of Pre-clinical Innovation, National Center for
Advancing Translational Sciences, National Institutes of Health, 9800 Medical
Center Drive, Rockville, Maryland
20850, USA
| | - Sampada A. Shahane
- Division of Pre-clinical Innovation, National Center for
Advancing Translational Sciences, National Institutes of Health, 9800 Medical
Center Drive, Rockville, Maryland
20850, USA
| | - Matias Attene-Ramos
- Division of Pre-clinical Innovation, National Center for
Advancing Translational Sciences, National Institutes of Health, 9800 Medical
Center Drive, Rockville, Maryland
20850, USA
| | - Tongan Zhao
- Division of Pre-clinical Innovation, National Center for
Advancing Translational Sciences, National Institutes of Health, 9800 Medical
Center Drive, Rockville, Maryland
20850, USA
| | - Christopher P. Austin
- Division of Pre-clinical Innovation, National Center for
Advancing Translational Sciences, National Institutes of Health, 9800 Medical
Center Drive, Rockville, Maryland
20850, USA
| | - Anton Simeonov
- Division of Pre-clinical Innovation, National Center for
Advancing Translational Sciences, National Institutes of Health, 9800 Medical
Center Drive, Rockville, Maryland
20850, USA
| |
Collapse
|
24
|
Kutty RV, Chia SL, Setyawati MI, Muthu MS, Feng SS, Leong DT. In vivo and ex vivo proofs of concept that cetuximab conjugated vitamin E TPGS micelles increases efficacy of delivered docetaxel against triple negative breast cancer. Biomaterials 2015; 63:58-69. [DOI: 10.1016/j.biomaterials.2015.06.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Revised: 06/03/2015] [Accepted: 06/03/2015] [Indexed: 01/31/2023]
|
25
|
Fagan-Solis KD, Reaves DK, Rangel MC, Popoff MR, Stiles BG, Fleming JM. Challenging the roles of CD44 and lipolysis stimulated lipoprotein receptor in conveying Clostridium perfringens iota toxin cytotoxicity in breast cancer. Mol Cancer 2014; 13:163. [PMID: 24990559 PMCID: PMC4086999 DOI: 10.1186/1476-4598-13-163] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 06/24/2014] [Indexed: 01/01/2023] Open
Abstract
Background Translational exploration of bacterial toxins has come to the forefront of research given their potential as a chemotherapeutic tool. Studies in select tissues have demonstrated that Clostridium perfringens iota toxin binds to CD44 and lipolysis stimulated lipoprotein receptor (LSR) cell-surface proteins. We recently demonstrated that LSR expression correlates with estrogen receptor positive breast cancers and that LSR signaling directs aggressive, tumor-initiating cell behaviors. Herein, we identify the mechanisms of iota toxin cytotoxicity in a tissue-specific, breast cancer model with the ultimate goal of laying the foundation for using iota toxin as a targeted breast cancer therapy. Methods In vitro model systems were used to determine the cytotoxic effect of iota toxin on breast cancer intrinsic subtypes. The use of overexpression and knockdown technologies confirmed the roles of LSR and CD44 in regulating iota toxin endocytosis and induction of cell death. Lastly, cytotoxicity assays were used to demonstrate the effect of iota toxin on a validated set of tamoxifen resistant breast cancer cell lines. Results Treatment of 14 breast cancer cell lines revealed that LSR+/CD44- lines were highly sensitive, LSR+/CD44+ lines were slightly sensitive, and LSR-/CD44+ lines were resistant to iota cytotoxicity. Reduction in LSR expression resulted in a significant decrease in toxin sensitivity; however, overexpression of CD44 conveyed toxin resistance. CD44 overexpression was correlated with decreased toxin-stimulated lysosome formation and decreased cytosolic levels of iota toxin. These findings indicated that expression of CD44 drives iota toxin resistance through inhibition of endocytosis in breast cancer cells, a role not previously defined for CD44. Moreover, tamoxifen-resistant breast cancer cells exhibited robust expression of LSR and were highly sensitive to iota-induced cytotoxicity. Conclusions Collectively, these data are the first to show that iota toxin has the potential to be an effective, targeted therapy for breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Jodie M Fleming
- Department of Biology, North Carolina Central University, Durham, NC, USA.
| |
Collapse
|
26
|
Abstract
The majority of human breast cancers are estrogen receptor-positive (ER+), but this has proven challenging to model in genetically engineered mice. This review summarizes information on 21 mouse models that develop ER+ mammary cancer. Where available, information on cancer pathology and gene expression profiles is referenced to assist in understanding which histological subtype of ER+ human cancer each model might represent. ESR1, CCDN1, prolactin, TGFα, AIB1, ESPL1, and WNT1 overexpression, PIK3CA gain of function, as well as loss of P53 (Trp53) or STAT1 are associated with ER+ mammary cancer. Treatment with the PPARγ agonist efatutazone in a mouse with Brca1 and p53 deficiency and 7,12-dimethylbenz(a)anthracene exposure in combination with an activated myristoylated form of AKT1 also induce ER+ mammary cancer. A spontaneous mutant in nude mice that develops metastatic ER+ mammary cancer is included. Age of cancer development ranges from 3 to 26 months and the percentage of cancers that are ER+ vary from 21 to 100%. Not all models are characterized as to their estrogen dependency and/or response to anti-hormonal therapy. Strain backgrounds include C57Bl/6, FVB, BALB/c, 129S6/SvEv, CB6F1, and NIH nude. Most models have only been studied on one strain background. In summary, while a range of models are available for studies of pathogenesis and therapy of ER+ breast cancers, many could benefit from further characterization, and opportunity for development of new models remains.
Collapse
Affiliation(s)
- Sarah A. Dabydeen
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA 20057
| | - Priscilla A. Furth
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA 20057
- Department of Medicine, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA 20057
| |
Collapse
|
27
|
Godbole AM, Ramalingam S, Ramamurthy VP, Khandelwal A, Bruno RD, Upreti VV, Gediya LK, Purushottamachar P, Mbatia HW, Addya S, Ambulos N, Njar VCO. VN/14-1 induces ER stress and autophagy in HP-LTLC human breast cancer cells and has excellent oral pharmacokinetic profile in female Sprague Dawley rats. Eur J Pharmacol 2014; 734:98-104. [PMID: 24726842 DOI: 10.1016/j.ejphar.2014.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 12/19/2022]
Abstract
Resistance to aromatase inhibitors is a major concern in the treatment of breast cancer. Long-term letrozole cultured (LTLC) cells represent a model of resistance to aromatase inhibitors. The LTLC cells were earlier generated by culturing MCF-7Ca, the MCF-7 human breast cancer cell line stably transfected with human placental aromatase gene for a prolonged period in the presence of letrozole. In the present study the effect of RAMBA, VN/14-1 on the sensitivity of LTLC cells upon multiple passaging and the mechanisms of action of VN/14-1 in such high passage LTLC (HP-LTLC) cells was investigated. We report that multiple passaging of LTLC cells (HP-LTLC cell clones) led to profound decrease in their sensitivity to VN/14-1. Additionally, microarray studies and protein analysis revealed that VN/14-1 induced marked endoplasmic reticulum (ER) stress and autophagy in HP-LTLC cells. We further report that VN/14-1 in combination with thapsigargin exhibited synergistic anti-cancer effect in HP-LTLC cells. Preliminary pharmacokinetics in rats revealed that VN/14-1 reached a peak plasma concentration (Cmax) within 0.17h after oral dosing. Its absolute oral bioavailability was >100%. Overall these results indicate potential of VN/14-1 for further clinical development as a potential oral agent for the treatment of breast cancer.
Collapse
Affiliation(s)
- Abhijit M Godbole
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Senthilmurugan Ramalingam
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Vidya P Ramamurthy
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Aakanksha Khandelwal
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Robert D Bruno
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Vijay V Upreti
- Department of Pharmaceutical sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201, USA
| | - Lalji K Gediya
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Puranik Purushottamachar
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Hannah W Mbatia
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Sankar Addya
- Department of Cancer Biology, Kimmel Cancer Center, Jefferson Medical College, Thomas Jefferson University, PA 19107, USA
| | - Nicholas Ambulos
- Genomic Core Facility, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Vincent C O Njar
- Department of Pharmacology, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA; Marlene Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, 685 West Baltimore Street, Baltimore, MD 21201-1559, USA.
| |
Collapse
|
28
|
Kutty RV, Feng SS. Cetuximab conjugated vitamin E TPGS micelles for targeted delivery of docetaxel for treatment of triple negative breast cancers. Biomaterials 2013; 34:10160-71. [DOI: 10.1016/j.biomaterials.2013.09.043] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 09/13/2013] [Indexed: 12/28/2022]
|
29
|
Bouker KB, Wang Y, Xuan J, Clarke R. Antiestrogen Resistance and the Application of Systems Biology. ACTA ACUST UNITED AC 2012; 9:e11-e17. [PMID: 23539064 DOI: 10.1016/j.ddmec.2012.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Understanding the molecular changes that drive an acquired antiestrogen resistance phenotype is of major clinical relevance. Previous methodologies for addressing this question have taken a single gene/pathway approach and the resulting gains have been limited in terms of their clinical impact. Recent systems biology approaches allow for the integration of data from high throughput "-omics" technologies. We highlight recent advances in the field of antiestrogen resistance with a focus on transcriptomics, proteomics and methylomics.
Collapse
Affiliation(s)
- Kerrie B Bouker
- Department of Oncology and Lombardi Comprehensive Cancer Center, Georgetown University School of Medicine, Washington, DC 20057, U.S.A
| | | | | | | |
Collapse
|
30
|
Kubo M, Kanaya N, Petrossian K, Ye J, Warden C, Liu Z, Nishimura R, Osako T, Okido M, Shimada K, Takahashi M, Chu P, Yuan YC, Chen S. Inhibition of the proliferation of acquired aromatase inhibitor-resistant breast cancer cells by histone deacetylase inhibitor LBH589 (panobinostat). Breast Cancer Res Treat 2012; 137:93-107. [PMID: 23160924 DOI: 10.1007/s10549-012-2332-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 10/31/2012] [Indexed: 11/27/2022]
Abstract
Aromatase inhibitors (AIs) are important drugs for treating postmenopausal patients with hormone receptor-positive breast cancer. However, acquired resistance to AI therapies is a significant problem. Our study has revealed that the histone deacetylase inhibitor LBH589 treatment abrogated growth of AI-resistant cells in vitro and in vivo, causing cell cycle G2/M arrest and induced apoptosis. LBH589 treatment also reduced the level of NF-κB1 which is overexpressed when AI resistance develops. Analyzing paired tumor specimens from 12 patients, we found that NF-κB1 expression was increased in recurrent AI-resistant tumors as compared to the paired primary tumors before AI treatment. This finding was consistent with up-regulated NF-κB1 expression seen in a collection of well-established AI-resistant cell lines. Furthermore, knockdown of NF-κB1 expression significantly suppressed the proliferation of AI-resistant cells. Treatment of AI-resistant cell lines with LBH589 suppressed NF-κB1 mRNA and protein expression. In addition, LBH589 treatment abrogated growth of AI-resistant tumors in mice, and was associated with significantly decreased levels of NF-κB1 in tumors. In all, our findings strongly support further investigation of LBH589 as a novel therapeutic strategy for patients with AI-resistant breast cancer, in part by suppressing the NF-κB1 pathway.
Collapse
Affiliation(s)
- Makoto Kubo
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|