1
|
Verhoog NJD, Spies LML. The anti-aromatase and anti-estrogenic activity of plant products in the treatment of estrogen receptor-positive breast cancer. J Steroid Biochem Mol Biol 2024; 243:106581. [PMID: 38997071 DOI: 10.1016/j.jsbmb.2024.106581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 06/14/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Despite being the focal point of decades of research, female breast cancer (BC) continues to be one of the most lethal cancers in the world. Given that 80 % of all diagnosed BC cases are estrogen receptor-positive (ER+) with carcinogenesis driven by estrogen-ERα signalling, current standard of care (SOC) hormone therapies are geared towards modulating the function and expression levels of estrogen and its receptors, ERα and ERβ. Currently, aromatase inhibitors (AIs), selective ER modulators (SERMs) and selective ER degraders (SERDs) are clinically prescribed for the management and treatment of ER+ BC, with the anti-aromatase activity of AIs abrogating estrogen biosynthesis, while the anti-estrogenic SERMs and SERDs antagonise and degrade the ER, respectively. The use of SOC hormone therapies is, however, significantly hampered by the onset of severe side-effects and the development of resistance. Given that numerous studies have reported on the beneficial effects of plant compounds and/or extracts and the multiple pathways through which they target ER+ breast carcinogenesis, recent research has focused on the use of dietary chemopreventive agents for BC management. When combined with SOC treatments, several of these plant components and/or extracts have demonstrated improved efficacy and/or synergistic impact. Moreover, despite a lack of in vivo investigations, plant products are generally reported to have a lower side-effect profile than SOC therapies and are therefore thought to be a safer therapeutic choice. Thus, the current review summarizes the findings from the last five years regarding the anti-aromatase and anti-estrogenic activity of plant products, as well as their synergistic anti-ER+ BC effects in combination with SOC therapies.
Collapse
Affiliation(s)
| | - Lee-Maine Lorin Spies
- Department of Biochemistry, Stellenbosch University, Van de Byl Street, Stellenbosch, 7601, South Africa
| |
Collapse
|
2
|
Lee SJ, Cha CD, Hong H, Choi YY, Chung MS. Adverse effects of tamoxifen treatment on bone mineral density in premenopausal patients with breast cancer: a systematic review and meta-analysis. Breast Cancer 2024; 31:717-725. [PMID: 38671211 DOI: 10.1007/s12282-024-01586-2] [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: 08/03/2023] [Accepted: 04/11/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND It is well known that adjuvant tamoxifen treatment for breast cancer in postmenopausal women decreased bone loss. However, the effects of adjuvant tamoxifen therapy on bone mineral density (BMD) in premenopausal patients with breast cancer remains uncertain. Tamoxifen would have a potential impact of premenopausal BMD on health. The aim of this meta-analysis was to assess this in premenopausal women with primary breast cancer. METHODS Through April 2020, studies reporting BMD changes of lumbar spine or hip in premenopausal women with primary breast cancer treated with adjuvant tamoxifen and tamoxifen plus chemotherapy or ovarian function suppression (OFS) were collected from EMBASE and PubMed. The meta-analysis was performed using random effects model of the standardized mean difference (SMD) of BMD in patients. RESULTS A total of 1432 premenopausal patients were enrolled in eight studies, involving 198 patients treated with tamoxifen alone in three studies. After a 3-year median follow-up, adjuvant tamoxifen decreased the lumbar spinal and hip BMD by as much as an SMD of -1.17 [95% confidence interval (CI); -1.58 to -0.76)] and -0.66 (95% CI, -1.55 to 0.23), respectively. In subgroup analysis in patients treated adjuvant tamoxifen and tamoxifen plus chemotherapy or OFS according to follow-up duration, the bone change of < 3 years follow-up group was -0.03 SMD (95% CI, -0.47 to 0.41) and that of ≥ 3 years follow-up group was -1.06 SMD (95% CI, -1.48 to -0.64). Compared with patients who received tamoxifen alone, patients who received combination therapy with chemotherapy or OFS showed lesser bone loss at the lumbar spine. CONCLUSIONS Our meta-analysis demonstrated that adjuvant tamoxifen therapy in premenopausal patients caused bone loss after 3 years of follow-up, especially at the lumbar spines. For a definite evaluation of the adverse effects of tamoxifen on bone, it is necessary to accumulate more relevant studies.
Collapse
Affiliation(s)
- Soo Jin Lee
- Department of Nuclear Medicine, Hanyang University Medical Center, Hanyang University College of Medicine, 222-1 Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, South Korea
| | - Chihwan David Cha
- Department of Surgery, Hanyang University Medical Center, Hanyang University College of Medicine, 222-1 Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, South Korea
| | - Hanpyo Hong
- Biostatistical Consulting and Research Lab, Medical Research Collaborating Center, Hanyang University, 222 Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, South Korea
| | - Yun Young Choi
- Department of Nuclear Medicine, Hanyang University Medical Center, Hanyang University College of Medicine, 222-1 Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, South Korea
| | - Min Sung Chung
- Department of Surgery, Hanyang University Medical Center, Hanyang University College of Medicine, 222-1 Wangsimni-Ro, Seongdong-Gu, Seoul, 04763, South Korea.
| |
Collapse
|
3
|
Cucciniello L, Garufi G, Di Rienzo R, Martinelli C, Pavone G, Giuliano M, Arpino G, Montemurro F, Del Mastro L, De Laurentiis M, Puglisi F. Estrogen deprivation effects of endocrine therapy in breast cancer patients: Incidence, management and outcome. Cancer Treat Rev 2023; 120:102624. [PMID: 37751658 DOI: 10.1016/j.ctrv.2023.102624] [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: 06/23/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023]
Abstract
Endocrine therapy is one of the standard adjuvant treatments to reduce the risk of recurrence and mortality in patients with hormone receptor positive early breast cancer. Despite its proven efficacy, ET side effects, which persist over time even if low grade, may deteriorate quality of life. During follow-up visits, emphasis is generally placed on the risk of disease recurrence, while the topic of ET side effects is commonly neglected and discussed only briefly. This could lead to poor adherence to therapy and early treatment discontinuation, resulting in worse survival outcomes. The aim of this review is to provide an overview of the available evidence on the incidence and reporting of ET-related side effects (including vasomotor symptoms, musculoskeletal disorders and genitourinary syndrome of menopause, as well as fatigue, psychological and ocular disorders, dysmetabolic effects and loss of bone density) and of the pharmacological and non-pharmacological strategies available to mitigate symptom burden.
Collapse
Affiliation(s)
- Linda Cucciniello
- Department of Medicine, University of Udine, Udine, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy.
| | - Giovanna Garufi
- Department of Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy; Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, IRCCS, Rome, Italy.
| | - Rossana Di Rienzo
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Claudia Martinelli
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Giuliana Pavone
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy; Center of Experimental Oncology and Hematology, A.O.U. Policlinico "G. Rodolico - San Marco", Catania, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy.
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | | | - Lucia Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy; Breast Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | | | - Fabio Puglisi
- Department of Medicine, University of Udine, Udine, Italy; Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy.
| |
Collapse
|
4
|
Nahmias-Blank D, Maimon O, Meirovitz A, Sheva K, Peretz-Yablonski T, Elkin M. Excess body weight and postmenopausal breast cancer: Emerging molecular mechanisms and perspectives. Semin Cancer Biol 2023; 96:26-35. [PMID: 37739109 DOI: 10.1016/j.semcancer.2023.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/24/2023]
Abstract
Postmenopausal, obese women have a significantly higher risk of developing estrogen receptor-positive (ER+) breast tumors, that are resistant to therapies and are associated with higher recurrence and death rates. The global prevalence of overweight/obese women has reached alarming proportions and with postmenopausal ER+ breast carcinoma (BC) having the highest incidence among the three obesity-related cancers in females (i.e., breast, endometrial and ovarian), this is of significant concern. Elucidation of the precise molecular mechanisms underlying the pro-cancerous action of obesity in ER+BC is therefore critical for disease prevention and novel treatment initiatives. Interestingly, accumulating data has shown opposing relationships between obesity and cancer in either pre- or post-menopausal women. Excess body weight is associated with an increased risk of breast cancer in postmenopausal women and a decreased risk in pre-menopausal women. Moreover, excess adiposity during early life appears to be protective against postmenopausal breast cancer, including both ER+ and ER negative BC subtypes. Overall, estrogen-dependent mechanisms have been implicated as the main driving force in obesity-related breast tumorigenesis. In the present review we discuss the epidemiologic and mechanistic aspects of association between obesity and breast tumors after menopause, mainly in the context of hormone dependency. Molecular and cellular events underlying this association present as potential avenues for both therapeutic intervention as well as the prevention of BC-promoting processes linked to excess adiposity, which is proving to be vital in an increasingly obese global population.
Collapse
Affiliation(s)
- Daniela Nahmias-Blank
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ofra Maimon
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amichay Meirovitz
- Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka University Medical Center, Be'er Sheva 84101, Israel
| | - Kim Sheva
- Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka University Medical Center, Be'er Sheva 84101, Israel
| | - Tamar Peretz-Yablonski
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; Hebrew University Medical School, Jerusalem 91120, Israel
| | - Michael Elkin
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel; Hebrew University Medical School, Jerusalem 91120, Israel.
| |
Collapse
|
5
|
Hammarström M, Gabrielson M, Crippa A, Discacciati A, Eklund M, Lundholm C, Bäcklund M, Wengström Y, Borgquist S, Bergqvist J, Eriksson M, Tapia J, Czene K, Hall P. Side effects of low-dose tamoxifen: results from a six-armed randomised controlled trial in healthy women. Br J Cancer 2023:10.1038/s41416-023-02293-z. [PMID: 37149701 DOI: 10.1038/s41416-023-02293-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Adherence to adjuvant tamoxifen therapy is suboptimal, and acceptance of tamoxifen for primary prevention is poor. Published results indicate effect of low-dose tamoxifen therapy. Using questionnaire data from a randomised controlled trial, we describe side effects of standard and low-dose tamoxifen in healthy women. METHODS In the KARISMA trial, 1440 healthy women were randomised to 6 months of daily intake of 20, 10, 5, 2.5, 1 mg of tamoxifen or placebo. Participants completed a 48-item, five-graded Likert score symptom questionnaire at baseline and follow-up. Linear regression models were used to identify significant changes in severity levels across doses and by menopausal status. RESULTS Out of 48 predefined symptoms, five were associated with tamoxifen exposure (hot flashes, night sweats, cold sweats, vaginal discharge and muscle cramps). When comparing these side effects in premenopausal women randomised to low doses (2.5, 5 mg) versus high doses (10, 20 mg), the mean change was 34% lower in the low-dose group. No dose-dependent difference was seen in postmenopausal women. CONCLUSIONS Symptoms related to tamoxifen therapy are influenced by menopausal status. Low-dose tamoxifen, in contrast to high-dose, was associated with less pronounced side effects, a finding restricted to premenopausal women. Our findings give new insights which may influence future dosing strategies of tamoxifen in both the adjuvant and preventive settings. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT03346200.
Collapse
Affiliation(s)
- Mattias Hammarström
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Marike Gabrielson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Alessio Crippa
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Andrea Discacciati
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Martin Eklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Cecilia Lundholm
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Bäcklund
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Yvonne Wengström
- Department of Neurobiology, Care Science and Society, Division of Nursing and Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Signe Borgquist
- Department of Oncology, Aarhus University Hospital and Aarhus University, Aarhus, Denmark
- Department of Clinical Sciences Lund, Oncology, Lund University and Skåne University Hospital, Lund, Sweden
| | - Jenny Bergqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Capio St Görans Hospital, Stockholm, Sweden
| | - Mikael Eriksson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - José Tapia
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Kamila Czene
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Per Hall
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Södersjukhuset, Stockholm, Sweden
| |
Collapse
|
6
|
Chakraborty B, Byemerwa J, Krebs T, Lim F, Chang CY, McDonnell DP. Estrogen Receptor Signaling in the Immune System. Endocr Rev 2023; 44:117-141. [PMID: 35709009 DOI: 10.1210/endrev/bnac017] [Citation(s) in RCA: 53] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Indexed: 01/14/2023]
Abstract
The immune system functions in a sexually dimorphic manner, with females exhibiting more robust immune responses than males. However, how female sex hormones affect immune function in normal homeostasis and in autoimmunity is poorly understood. In this review, we discuss how estrogens affect innate and adaptive immune cell activity and how dysregulation of estrogen signaling underlies the pathobiology of some autoimmune diseases and cancers. The potential roles of the major circulating estrogens, and each of the 3 estrogen receptors (ERα, ERβ, and G-protein coupled receptor) in the regulation of the activity of different immune cells are considered. This provides the framework for a discussion of the impact of ER modulators (aromatase inhibitors, selective estrogen receptor modulators, and selective estrogen receptor downregulators) on immunity. Synthesis of this information is timely given the considerable interest of late in defining the mechanistic basis of sex-biased responses/outcomes in patients with different cancers treated with immune checkpoint blockade. It will also be instructive with respect to the further development of ER modulators that modulate immunity in a therapeutically useful manner.
Collapse
Affiliation(s)
- Binita Chakraborty
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jovita Byemerwa
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Taylor Krebs
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA.,Known Medicine, Salt Lake City, UT 84108, USA
| | - Felicia Lim
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
7
|
van Dyk L, Verhoog NJD, Louw A. Combinatorial treatments of tamoxifen and SM6Met, an extract from Cyclopia subternata Vogel, are superior to either treatment alone in MCF-7 cells. Front Pharmacol 2022; 13:1017690. [PMID: 36210845 PMCID: PMC9535530 DOI: 10.3389/fphar.2022.1017690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Synergistic drug combinations are not only popular in antibiotic, anti-microbial, immune disease (i.e., AIDS) and viral infection studies, but has also gained traction in the field of cancer research as a multi-targeted approach. It has the potential to lower the doses needed of standard of care (SOC) therapeutic agents, whilst maintaining an effective therapeutic level. Lower dosages could ameliorate the fundamental problems such as drug resistance and metastasis associated with current SOC therapies. In the current study, we show that the combination of SM6Met with (2)-4-hydroxytamoxifen (4-OH-Tam, the active metabolite of tamoxifen) produces a strong synergistic effect in terms of inhibiting MCF7 ER-positive (ER+) breast cancer cell proliferation and that a 20 times lower dose of 4-OH-Tam in combination with SM6Met is required to produce the same inhibitory effect on cell proliferation as 4-OH-Tam on its own. Cell cycle analyses of the best combination ratios of SM6Met and 4-OH-Tam also suggests that the combination results in increased accumulation of cells in the S-phase and in the apoptotic phase. Moreover, the best combination ratio (20:1) of SM6Met with 4-OH-Tam displayed greater anti-metastatic potential in terms of inhibiting ER+ breast cancer cell migration, invasion, and colony formation than the SOC therapy alone, suggesting that SM6Met together with 4-OH-Tam could be a viable drug combination for not only delaying resistance and ameliorating the negative side-effects associated with current SOC therapies, like tamoxifen, but could also provide a novel, more affordable therapeutic alternative for treating or preventing ER+ breast cancer metastasis.
Collapse
|
8
|
Gai L, Sun W. Recent advances in estrogen receptor-targeted probes conjugated to BODIPY dyes. Steroids 2022; 183:109031. [PMID: 35381270 DOI: 10.1016/j.steroids.2022.109031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/19/2022] [Accepted: 03/31/2022] [Indexed: 02/06/2023]
Abstract
Estrogens, is a class of steroid hormones associated with the occurrence and development of breast cancer, that bind to estrogen receptors (ER). The development of BODIPY-based fluorescent ligands for the ER has continued to gain tremendous attention over the past 20 years. This review focuses on the synthesis methods, optical properties, and biological activity of BODIPY fluorescent probes conjugated to ER ligands. These will provide new strategy for designing fluorescent probes for targeting estrogen receptors.
Collapse
Affiliation(s)
- Linlin Gai
- Central Laboratory, Weifang People's Hospital, Weifang, Shandong 261041, PR China.
| | - Weice Sun
- Vascular Surgery, Weifang Traditional Chinese Hospital, Weifang, Shandong 261041, PR China
| |
Collapse
|
9
|
Serdaroğlu G, Şahin N, Üstün E, Tahir MN, Arıcı C, Gürbüz N, Özdemir İ. PEPPSI type complexes: Synthesis, x-ray structures, spectral studies, molecular docking and theoretical investigations. Polyhedron 2021. [DOI: 10.1016/j.poly.2021.115281] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
10
|
Kim CY, Kim YC, Oh JH, Kim MH. HOXA5 confers tamoxifen resistance via the PI3K/AKT signaling pathway in ER-positive breast cancer. J Cancer 2021; 12:4626-4637. [PMID: 34149926 PMCID: PMC8210559 DOI: 10.7150/jca.59740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022] Open
Abstract
Tamoxifen is a commonly used drug to treat estrogen receptor-positive patients with breast cancer. Despite the outstanding efficacy of tamoxifen, approximately one-third of patients develop resistance toward it, thereby presenting a therapeutic challenge. HOX genes may be involved in the acquisition of tamoxifen resistance. In this study, we identified HOXA5, a member of the HOX gene family, as a marker of tamoxifen resistance. Using ChIP assay, we found that HOXA5 expression was significantly overexpressed in tamoxifen-resistant MCF7 (TAMR) breast cancer cells because of reduced H3K27me3 binding. HOXA5 upregulation resulted in activation of the PI3K/AKT signaling cascade, which in turn, led to p53 and p21 reduction, ultimately making the TAMR cells less apoptotic. Furthermore, elevated HOXA5 expression resulted in breast cancer cells acquiring more mesenchymal-like and stem cell traits associated with aggressive breast cancer phenotypes. In conclusion, our results delineate a mechanism by which HOXA5 promotes tumorigenesis, cancer progression, and tamoxifen resistance in breast cancer cells.
Collapse
Affiliation(s)
- Clara Yuri Kim
- Department of Anatomy, Embryology Laboratory, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Yu Cheon Kim
- Department of Anatomy, Embryology Laboratory, Yonsei University College of Medicine, Seoul 03722, Korea
- Department of Anatomy, Graduate School of Medical Science, Bain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Ji Hoon Oh
- Department of Anatomy, Embryology Laboratory, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Myoung Hee Kim
- Department of Anatomy, Embryology Laboratory, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
11
|
Bhardwaj P, Brown KA. Obese Adipose Tissue as a Driver of Breast Cancer Growth and Development: Update and Emerging Evidence. Front Oncol 2021; 11:638918. [PMID: 33859943 PMCID: PMC8042134 DOI: 10.3389/fonc.2021.638918] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/12/2021] [Indexed: 12/24/2022] Open
Abstract
Obesity is an established risk factor for breast cancer growth and progression. A number of advances have been made in recent years revealing new insights into this link. Early events in breast cancer development involve the neoplastic transformation of breast epithelial cells to cancer cells. In obesity, breast adipose tissue undergoes significant hormonal and inflammatory changes that create a mitogenic microenvironment. Many factors that are produced in obesity have also been shown to promote tumorigenesis. Given that breast epithelial cells are surrounded by adipose tissue, the crosstalk between the adipose compartment and breast epithelial cells is hypothesized to be a significant player in the initiation and progression of breast cancer in individuals with excess adiposity. The present review examines this crosstalk with a focus on obese breast adipose-derived estrogen, inflammatory mediators and adipokines, and how they are mechanistically linked to breast cancer risk and growth through stimulation of oxidative stress, DNA damage, and pro-oncogenic transcriptional programs. Pharmacological and lifestyle strategies targeting these factors and their downstream effects are evaluated for feasibility and efficacy in decreasing the risk of obesity-induced breast epithelial cell transformation and consequently, breast cancer development.
Collapse
Affiliation(s)
- Priya Bhardwaj
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, United States
| | - Kristy A. Brown
- Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, United States
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
12
|
Behl T, Sharma A, Sharma L, Sehgal A, Zengin G, Brata R, Fratila O, Bungau S. Exploring the Multifaceted Therapeutic Potential of Withaferin A and Its Derivatives. Biomedicines 2020; 8:E571. [PMID: 33291236 PMCID: PMC7762146 DOI: 10.3390/biomedicines8120571] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022] Open
Abstract
Withaferin A (WA), a manifold studied, C28-steroidal lactone withanolide found in Withania somnifera. Given its unique beneficial effects, it has gathered attention in the era of modern science. Cancer, being considered a "hopeless case and the leading cause of death worldwide, and the available conventional therapies have many lacunae in the form of side effects. The poly pharmaceutical natural compound, WA treatment, displayed attenuation of various cancer hallmarks by altering oxidative stress, promoting apoptosis, and autophagy, inhibiting cell proliferation, reducing angiogenesis, and metastasis progression. The cellular proteins associated with antitumor pathways were also discussed. WA structural modifications attack multiple signal transduction pathways and enhance the therapeutic outcomes in various diseases. Moreover, it has shown validated pharmacological effects against multiple neurodegenerative diseases by inhibiting acetylcholesterinases and butyrylcholinesterases enzyme activity, antidiabetic activity by upregulating adiponectin and preventing the phosphorylation of peroxisome proliferator-activated receptors (PPARγ), cardioprotective activity by AMP-activated protein kinase (AMPK) activation and suppressing mitochondrial apoptosis. The current review is an extensive survey of various WA associated disease targets, its pharmacokinetics, synergistic combination, modifications, and biological activities.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Aditi Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh 173229, India; (A.S.); (L.S.)
| | - Lalit Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh 173229, India; (A.S.); (L.S.)
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University Campus, Konya 42250, Turkey;
| | - Roxana Brata
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (R.B.); (O.F.)
| | - Ovidiu Fratila
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (R.B.); (O.F.)
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| |
Collapse
|
13
|
Sivasankarapillai VS, Madhu Kumar Nair R, Rahdar A, Bungau S, Zaha DC, Aleya L, Tit DM. Overview of the anticancer activity of withaferin A, an active constituent of the Indian ginseng Withania somnifera. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:26025-26035. [PMID: 32405942 DOI: 10.1007/s11356-020-09028-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/22/2020] [Indexed: 06/11/2023]
Abstract
Cancer is still considered a "hopeless case", besides all of the advancements in oncology research. On the other hand, the natural products, as effective lead molecules, have gained significant interest for research due to the absence of toxic and harmful side effects usually associated with conventional treatment methods. Medicinal properties of herbal plants are strongly evidenced in traditional medicine from ancient times. In the context above, withaferin A (WA) was identified as the active principle of the plant Withania somnifera, its molecule being reported to have excellent anticancer and tumour inhibition activities in various cell lines. Furthermore, the in silico approaches in the medicinal chemistry of WA revealed the biological targets and gave momentum for the research that leads to many amazing pharmacological activities of WA which are not yet explored. This includes a broad spectrum of anticancer actions manifested in different organs (breast, pancreas, colon), melanoma and B cell lymphoma, etc. This review is an extensive survey of the most recent anticancer studies reported for WA, along with its mechanism of action and details about its in vitro and/or in vivo behaviour.
Collapse
Affiliation(s)
| | | | - Abbas Rahdar
- Department of Physics, Faculty of Science,, University of Zabol, Zabol, Iran
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy,, University of Oradea, 410028, Oradea, Romania
| | - Dana Carmen Zaha
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy,, University of Oradea, 410028, Oradea, Romania
| | - Lotfi Aleya
- Laboratoire Chrono-environnement CNRS 6249, Université de Franche-Comté, Besançon, France.
| | - Delia Mirela Tit
- Department of Pharmacy, Faculty of Medicine and Pharmacy,, University of Oradea, 410028, Oradea, Romania
| |
Collapse
|
14
|
|
15
|
Kim CY, Oh JH, Lee JY, Kim MH. The LncRNA HOTAIRM1 Promotes Tamoxifen Resistance by Mediating HOXA1 Expression in ER+ Breast Cancer Cells. J Cancer 2020; 11:3416-3423. [PMID: 32284737 PMCID: PMC7150441 DOI: 10.7150/jca.38728] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 02/26/2020] [Indexed: 01/16/2023] Open
Abstract
Breast cancer is one of the most commonly diagnosed cancers in women worldwide. Approximately 40% of patients with breast cancer acquire endocrine resistance following therapy with tamoxifen. Many explanations for the development of endocrine resistance have been put forward, one of them being the dysregulation of long non-coding RNAs (lncRNAs). The lncRNA HOTAIRM1, known to be involved in myelopoiesis as well as transcriptional regulation of the HOXA genes in embryonic stem cells, is also expressed in breast cancer cells. This study explored the molecular mechanisms of HOTAIRM1 involved in acquired tamoxifen resistance. We showed that HOTAIRM1 and HOXA1 are concurrently up-regulated in tamoxifen-resistant MCF7 (TAMR) cells. Knockdown of HOTAIRM1 down-regulated HOXA1 expression and restored sensitivity to tamoxifen. In addition, the knockdown of HOXA1 showed similar effects, suggesting that the HOTAIRM1/HOXA1 axis regulates tamoxifen resistance. Furthermore, we showed that HOTAIRM1 directly interacts with EZH2 and prevents the PRC2 complex from binding and depositing H3K27me3 on the putative promoter of HOXA1. Together, our findings suggest that HOXA1 and its neighboring lncRNA, HOTAIRM1, might serve as potential therapeutic targets for ER+ breast cancer patients who have acquired tamoxifen resistance.
Collapse
Affiliation(s)
- Clara Yuri Kim
- Department of Anatomy, Embryology Laboratory, Yonsei University College of Medicine, Seoul 03722, Korea.,Brain Korea 21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Ji Hoon Oh
- Department of Anatomy, Embryology Laboratory, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Ji-Yeon Lee
- Department of Anatomy, Embryology Laboratory, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Myoung Hee Kim
- Department of Anatomy, Embryology Laboratory, Yonsei University College of Medicine, Seoul 03722, Korea.,Brain Korea 21 PLUS project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
16
|
Cheung YM, Ramchand SK, Yeo B, Grossmann M. Cardiometabolic Effects of Endocrine Treatment of Estrogen Receptor-Positive Early Breast Cancer. J Endocr Soc 2019; 3:1283-1301. [PMID: 31259291 PMCID: PMC6595530 DOI: 10.1210/js.2019-00096] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022] Open
Abstract
Estrogen receptor-positive early breast cancer is common and has a relatively good prognosis. It shares risk factors with cardiovascular disease, and cardiovascular disease is an important competing cause of mortality. Adjuvant endocrine therapy with aromatase inhibitors (requiring concomitant ovarian suppression in premenopausal women) or selective estrogen receptor modulators (usually tamoxifen) exert oncologic benefits by respectively inhibiting estradiol synthesis or breast estrogen receptor signaling. Aromatase inhibitors cause systemic estradiol depletion. Tamoxifen has mixed agonistic/antagonistic effects in a tissue-dependent fashion. Given that estrogens modulate cardiometabolic risk, a review of the effects of endocrine therapy on cardiometabolic outcomes is pertinent. The current, but limited, evidence suggests that tamoxifen treatment, although associated with increases in body fat, hepatic steatosis, serum triglycerides, and diabetes risk, modestly reduces low-density lipoprotein cholesterol and lipoprotein(a) and may have favorable effects on markers of subclinical atherosclerosis. Tamoxifen is associated with either no effect on, or a reduction in, cardiovascular events, and it is associated with an increase in venous thromboembolic events. Aromatase inhibitors, although fewer studies are available and often confounded by comparison with tamoxifen, have not been consistently associated with adverse changes in cardiometabolic risk factors or increases in cardiovascular events. Further clinical trials designed to evaluate cardiometabolic outcomes are needed to more accurately determine the effects of endocrine therapy on cardiovascular risks, to inform individualized decisions regarding choice and duration of endocrine therapy, and to implement evidence-based strategies to mitigate cardiometabolic risks. In the meantime, although breast cancer-specific evidence for benefit of lifestyle measures is available and recommended routinely, proactive monitoring and treatment of cardiovascular risk factors should follow general population recommendations.
Collapse
Affiliation(s)
- Yee-Ming Cheung
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria Australia
| | - Sabashini K Ramchand
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria Australia
| | - Belinda Yeo
- Department of Medical Oncology, Austin Health, Heidelberg, Victoria, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
| | - Mathis Grossmann
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, Australia
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, Victoria Australia
| |
Collapse
|
17
|
El-Boubbou K, Ali R, Al-Zahrani H, Trivilegio T, Alanazi AH, Khan AL, Boudjelal M, AlKushi A. Preparation of iron oxide mesoporous magnetic microparticles as novel multidrug carriers for synergistic anticancer therapy and deep tumor penetration. Sci Rep 2019; 9:9481. [PMID: 31263250 PMCID: PMC6603044 DOI: 10.1038/s41598-019-46007-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 06/18/2019] [Indexed: 12/13/2022] Open
Abstract
The preparation of mesoporous iron oxides with controllable physiochemical properties for effective therapeutic drug delivery remains a formidable challenge. Herein, iron oxide mesoporous magnetic microparticles (IO-MMMs) were prepared by a modified reverse hard-templating approach using, for the first time, acid-prepared mesoporous spheres (APMS) as the hard silica template. The obtained mesostructures exhibited remarkably high surface area and large pore volumes (SBET = 240 m2/g and Vpore = 0.55 cm3/g), controllable average sizes, generally uniform morphologies, and excellent biocompatibilities, allowing them to achieve optimal drug release in cancer cells and tumor tissues. IO-MMM carriers were able to co-load high amounts of hydrophilic chemotherapeutic drugs (Dox or Daun) and/or hydrophobic hormonal anticancer drugs (Tam), and release them sustainably in a pH-dependent manner, utilizing the fluorescence of Daun to real-time trace the intracellular drug distribution, and employing Daun/Tam to treat cancer by combined chemo/hormonal therapy. Cytotoxicity assays against different types of cancerous cells showed that the combinatory Daun/Tam@IO-MMM formulation significantly reduced the viability of metastatic MCF7 and KAIMRC1 breast as well as HCT8 colorectal cancer cells, with the least potency towards non-cancerous normal primary cells (up to 10-fold). Electron, flow, and live confocal microscopy imaging confirmed that the loaded vehicles were successfully and differentially uptaken by the different tested cells, gradually releasing their payloads, and causing apoptotic cell death. Importantly, compared to free drugs, Daun/Tam@IO-MMMs displayed enhanced drug accumulation in patient breast primary tumor tissues, deeply penetrating into the tumor region and killing the tumor cells inside. The designed carriers described here, thus, constitute a novel promising magnetic mesoporous smart system that entraps different kinds of drugs and release them in a controlled manner for combinatorial chemo/hormonal cancer theranostics. This multifactorial platform may open new avenues in cancer therapy as efficient synergistic antitumor system through overcoming limitations of conventional cancer therapy.
Collapse
Affiliation(s)
- Kheireddine El-Boubbou
- Department of Basic Sciences, College of Science & Health Professions (COSHP), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City, National Guard Health Affairs, Riyadh, 11481, Saudi Arabia. .,King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City, National Guard Health Affairs, Riyadh, 11426, Saudi Arabia.
| | - Rizwan Ali
- King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City, National Guard Health Affairs, Riyadh, 11426, Saudi Arabia
| | - Hajar Al-Zahrani
- King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City, National Guard Health Affairs, Riyadh, 11426, Saudi Arabia
| | - Thadeo Trivilegio
- King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City, National Guard Health Affairs, Riyadh, 11426, Saudi Arabia
| | - Abdullah H Alanazi
- Department of Pathology and Laboratory Medicine, King Abdulaziz Medical City, National Guard Health Affairs, Riyadh, 11426, Saudi Arabia
| | - Abdul Latif Khan
- Department of Pathology and Laboratory Medicine, King Abdulaziz Medical City, National Guard Health Affairs, Riyadh, 11426, Saudi Arabia
| | - Mohamed Boudjelal
- King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City, National Guard Health Affairs, Riyadh, 11426, Saudi Arabia
| | - Abdulmohsen AlKushi
- Department of Basic Sciences, College of Science & Health Professions (COSHP), King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City, National Guard Health Affairs, Riyadh, 11481, Saudi Arabia
| |
Collapse
|
18
|
Linares A, Assou S, Lapierre M, Thouennon E, Duraffourd C, Fromaget C, Boulahtouf A, Tian G, Ji J, Sahin O, Badia E, Boulle N, Cavaillès V. Increased expression of the HDAC9 gene is associated with antiestrogen resistance of breast cancers. Mol Oncol 2019; 13:1534-1547. [PMID: 31099456 PMCID: PMC6599838 DOI: 10.1002/1878-0261.12505] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 04/26/2019] [Accepted: 05/15/2019] [Indexed: 12/19/2022] Open
Abstract
Estrogens play a pivotal role in breast cancer etiology, and endocrine therapy remains the main first line treatment for estrogen receptor‐alpha (ERα)‐positive breast cancer. ER are transcription factors whose activity is finely regulated by various regulatory complexes, including histone deacetylases (HDACs). Here, we investigated the role of HDAC9 in ERα signaling and response to antiestrogens in breast cancer cells. Various Michigan Cancer Foundation‐7 (MCF7) breast cancer cell lines that overexpress class IIa HDAC9 or that are resistant to the partial antiestrogen 4‐hydroxy‐tamoxifen (OHTam) were used to study phenotypic changes in response to ER ligands by using transcriptomic and gene set enrichment analyses. Kaplan–Meier survival analyses were performed using public transcriptomic datasets from human breast cancer biopsies. In MCF7 breast cancer cells, HDAC9 decreased ERα mRNA and protein expression and inhibited its transcriptional activity. Conversely, HDAC9 mRNA was strongly overexpressed in OHTam‐resistant MCF7 cells and in ERα‐negative breast tumor cell lines. Moreover, HDAC9‐overexpressing cells were less sensitive to OHTam antiproliferative effects compared with parental MCF7 cells. Several genes (including MUC1, SMC3 and S100P) were similarly deregulated in OHTam‐resistant and in HDAC9‐overexpressing MCF7 cells. Finally, HDAC9 expression was positively associated with genes upregulated in endocrine therapy‐resistant breast cancers and high HDAC9 levels were associated with worse prognosis in patients treated with OHTam. These results demonstrate the complex interactions of class IIa HDAC9 with ERα signaling in breast cancer cells and its effect on the response to hormone therapy.
Collapse
Affiliation(s)
- Aurélien Linares
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Said Assou
- Université Montpellier, France.,IRMB, Institute for Regenerative Medicine & Biotherapy, Montpellier, France.,INSERM, U1183, Montpellier, France
| | - Marion Lapierre
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Erwan Thouennon
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Céline Duraffourd
- Laboratoire de Biopathologie des Tumeurs, CHU Arnaud de Villeneuve, Montpellier, France
| | - Carole Fromaget
- Laboratoire de Biopathologie des Tumeurs, CHU Arnaud de Villeneuve, Montpellier, France
| | - Abdelhay Boulahtouf
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Gao Tian
- Key Laboratory of Carcinogenesis and Translational Research Ministry of Education, Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research Ministry of Education, Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ozgur Sahin
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC, USA
| | - Eric Badia
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Nathalie Boulle
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France.,Laboratoire de Biopathologie des Tumeurs, CHU Arnaud de Villeneuve, Montpellier, France
| | - Vincent Cavaillès
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| |
Collapse
|
19
|
Jia X, Li C, Li L, Liu X, Zhou L, Zhang W, Ni S, Lu Y, Chen L, Jeong LS, Yu J, Zhang Y, Zhang J, He S, Hu X, Sun H, Yu K, Liu G, Zhao H, Zhang Y, Jia L, Shao ZM. Neddylation Inactivation Facilitates FOXO3a Nuclear Export to Suppress Estrogen Receptor Transcription and Improve Fulvestrant Sensitivity. Clin Cancer Res 2019; 25:3658-3672. [PMID: 30833270 DOI: 10.1158/1078-0432.ccr-18-2434] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 01/04/2019] [Accepted: 02/28/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE How the neddylation pathway functions in breast tumor and regulation of estrogen receptor (ER) expression is rarely reported. The purpose of this study was to identify the role of neddylation in breast cancer and ER expression, and further explore the underlying mechanisms. EXPERIMENTAL DESIGN Expression patterns of nedd8-activating enzyme (NAE) and nedd8, two key proteins in the neddylation pathway, were examined in human breast specimens. ER-α expression was investigated using animal 18F-FES-PET/CT and immunoblotting upon NAE inhibitor MLN4924 treatment. Chromatin immunoprecipitation assay, luciferase reporter promoter assay, and the CRISPR-Cas9 system were used to elucidate the mechanism of ER-α regulation by MLN4924. The ER-positive breast cancer mouse model was used to determine the synergetic effect of MLN4924 and fulvestrant on tumor growth. All statistical tests were two-sided. RESULTS Both NAE1 and nedd8 expressions were higher in the ER-positive subgroup. Higher expressions of NAE1 and nedd8 indicated poorer prognosis. Importantly, ER-α expression was significantly downregulated upon MLN4924 treatment in vitro and in vivo. Mechanistically, MLN4924 treatment delayed serum and glucocorticoid-induced protein kinase (SGK) degradation and induced Forkhead box O3a (FOXO3a) nuclear export as well as decreased binding to the ESR1 promoter. Importantly, MLN4924 single or synergized with fulvestrant significantly suppressed the growth of ER-positive breast cancer in vitro and in vivo. CONCLUSIONS Our proof-of-principle study determines the activation of neddylation in breast tumor tissues for the first time and reveals a new ER-α regulatory mechanism, as well as further explores an effective approach to improve fulvestrant sensitivity through a neddylation inactivation combination.
Collapse
Affiliation(s)
- Xiaoqing Jia
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chunjie Li
- Department of Oncology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lihui Li
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoli Liu
- Cancer Institute, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lisha Zhou
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenjuan Zhang
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuaishuai Ni
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yun Lu
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Chen
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lak Shin Jeong
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Jinha Yu
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Yingjian Zhang
- Department of Nuclear Medicine, Center for Biomedical Imaging, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai, China
| | - Jianping Zhang
- Department of Nuclear Medicine, Center for Biomedical Imaging, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai, China
| | - Simin He
- Department of Nuclear Medicine, Center for Biomedical Imaging, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Engineering Research Center for Molecular Imaging Probes, Shanghai, China
| | - Xin Hu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hefen Sun
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Keda Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guangyu Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hu Zhao
- Department of Laboratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Yanmei Zhang
- Department of Laboratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai Key Laboratory of Clinical Geriatric Medicine, Research Center on Aging and Medicine, Fudan University, Shanghai, China
| | - Lijun Jia
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zhi-Ming Shao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, China. .,Department of Oncology, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Liu Z, Cheng Y, Luan Y, Zhong W, Lai H, Wang H, Yu H, Yang Y, Feng N, Yuan F, Huang R, He Z, Zhang F, Yan M, Yin H, Guo F, Zhai Q. Short-term tamoxifen treatment has long-term effects on metabolism in high-fat diet-fed mice with involvement of Nmnat2 in POMC neurons. FEBS Lett 2018; 592:3305-3316. [PMID: 30192985 DOI: 10.1002/1873-3468.13240] [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: 05/19/2018] [Revised: 08/16/2018] [Accepted: 08/25/2018] [Indexed: 01/07/2023]
Abstract
Short-term tamoxifen treatment has effects on lipid and glucose metabolism in mice fed chow. However, its effects on metabolism in mice fed high-fat diet (HFD) and the underlying mechanisms are unclear. Here, we show that tamoxifen treatment for 5 days decreases fat mass for as long as 18 weeks in mice fed HFD. Tamoxifen alters mRNA levels of some genes involved in lipid metabolism in white adipose tissue and improves glucose and insulin tolerance as well as hepatic insulin signaling for 12-20 weeks. Proopiomelanocortin (POMC) neuron-specific deletion of nicotinamide mononucleotide adenylyltransferase 2 (Nmnat2) attenuates the effects of tamoxifen on glucose and insulin tolerance. These data demonstrate that short-term injection of tamoxifen has long-term effects on lipid and glucose metabolism in HFD mice with involvement of Nmnat2 in POMC neurons.
Collapse
Affiliation(s)
- Zhiyuan Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yalan Cheng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yi Luan
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Wuling Zhong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Hejin Lai
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Hui Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Huimin Yu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Yale Yang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Ning Feng
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Feixiang Yuan
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Rui Huang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Zhishui He
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Fang Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Menghong Yan
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Hao Yin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Feifan Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Qiwei Zhai
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200031, Shanghai, China.,School of Life Science and Technology, Shanghai Tech University, Shanghai, 200093, China
| |
Collapse
|
21
|
Jose A, Ninave KM, Karnam S, Venuganti VVK. Temperature-sensitive liposomes for co-delivery of tamoxifen and imatinib for synergistic breast cancer treatment. J Liposome Res 2018; 29:153-162. [PMID: 30022700 DOI: 10.1080/08982104.2018.1502315] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Co-delivery of chemotherapeutic agents using nanocarriers is a promising strategy for enhancing therapeutic efficacy of anticancer agents. The aim of this work was to develop tamoxifen and imatinib dual drug loaded temperature-sensitive liposomes to treat breast cancer. Liposomes were prepared using 1, 2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), monopalmitoyl-2-hydroxy-sn-glycero-3-phosphocholine (MPPC), and different surface active agents. The liposomes were characterized for the average particle size, zeta potential, transition temperature, and drug release below and above liposomal transition temperature. The temperature-sensitive liposomes co-encapsulated with tamoxifen and imatinib were investigated for their synergistic activity against MCF-7 and MDA-MB-231 breast cancer cells. The liposomal nanoparticles showed a transition temperature of 39.4 °C and >70% encapsulation efficiency for tamoxifen and imatinib. The temperature-responsive liposomes showed more than 80% drug released within 30 min above transition temperature. Dual drug loaded liposomes showed synergistic growth inhibition against MCF-7 and MDA-MB-231 breast cancer cells. Co-delivery of tamoxifen and imatinib using temperature-sensitive liposomes can be developed as a potential targeting strategy against breast cancer.
Collapse
Affiliation(s)
- Anup Jose
- a Department of Pharmacy , Birla Institute of Technology and Science (BITS) Pilani , Hyderabad Campus , Hyderabad , India
| | - Kunal Manoj Ninave
- a Department of Pharmacy , Birla Institute of Technology and Science (BITS) Pilani , Hyderabad Campus , Hyderabad , India
| | - Sriravali Karnam
- a Department of Pharmacy , Birla Institute of Technology and Science (BITS) Pilani , Hyderabad Campus , Hyderabad , India
| | | |
Collapse
|
22
|
Kaymak A, Sayols S, Papadopoulou T, Richly H. Role for the transcriptional activator ZRF1 in early metastatic events in breast cancer progression and endocrine resistance. Oncotarget 2018; 9:28666-28690. [PMID: 29983888 DOI: 10.18632/oncotarget.25596] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 05/24/2018] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is one of the most common malignancies among women which is often treated with hormone therapy and chemotherapy. Despite the improvements in detection and treatment of breast cancer, the vast majority of breast cancer patients are diagnosed with metastatic disease either at the beginning of the disease or later during treatment. Still, the molecular mechanisms causing a therapy resistant metastatic breast cancer are still elusive. In the present study we addressed the function of the transcriptional activator ZRF1 during breast cancer progression. We provide evidence that ZRF1 plays an essential role for the early metastatic events in vitro and acts like a tumor suppressor protein during the progression of breast invasive ductal carcinoma into a more advanced stage. Hence, depletion of ZRF1 results in the acquisition of metastatic behavior by facilitating the initiation of the metastatic cascade, notably for cell adhesion, migration and invasion. Furthermore absence of ZRF1 provokes endocrine resistance via misregulation of cell death and cell survival related pathways. Taken together, we have identified ZRF1 as an important regulator of breast cancer progression that holds the potential to be explored for new treatment strategies in the future.
Collapse
Affiliation(s)
- Aysegül Kaymak
- Laboratory of Molecular Epigenetics, Institute of Molecular Biology, Mainz, Germany
| | - Sergi Sayols
- Bioinformatics Core Facility, Institute of Molecular Biology, Mainz, Germany
| | - Thaleia Papadopoulou
- Laboratory of Molecular Epigenetics, Institute of Molecular Biology, Mainz, Germany.,Department of Developmental and Stem Cell Biology, Institute Pasteur, Paris, France
| | - Holger Richly
- Laboratory of Molecular Epigenetics, Institute of Molecular Biology, Mainz, Germany
| |
Collapse
|
23
|
Moradi N, Ashrafi-Kooshk MR, Chamani J, Shackebaei D, Norouzi F. Separate and simultaneous binding of tamoxifen and estradiol to human serum albumin: Spectroscopic and molecular modeling investigations. J Mol Liq 2018. [DOI: 10.1016/j.molliq.2017.11.056] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
24
|
Traboulsi T, El Ezzy M, Gleason JL, Mader S. Antiestrogens: structure-activity relationships and use in breast cancer treatment. J Mol Endocrinol 2017; 58:R15-R31. [PMID: 27729460 PMCID: PMC5148801 DOI: 10.1530/jme-16-0024] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 10/11/2016] [Indexed: 12/11/2022]
Abstract
About 70% of breast tumors express estrogen receptor alpha (ERα), which mediates the proliferative effects of estrogens on breast epithelial cells, and are candidates for treatment with antiestrogens, steroidal or non-steroidal molecules designed to compete with estrogens and antagonize ERs. The variable patterns of activity of antiestrogens (AEs) in estrogen target tissues and the lack of systematic cross-resistance between different types of molecules have provided evidence for different mechanisms of action. AEs are typically classified as selective estrogen receptor modulators (SERMs), which display tissue-specific partial agonist activity (e.g. tamoxifen and raloxifene), or as pure AEs (e.g. fulvestrant), which enhance ERα post-translational modification by ubiquitin-like molecules and accelerate its proteasomal degradation. Characterization of second- and third-generation AEs, however, suggests the induction of diverse ERα structural conformations, resulting in variable degrees of receptor downregulation and different patterns of systemic properties in animal models and in the clinic.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Hormonal/chemistry
- Antineoplastic Agents, Hormonal/pharmacology
- Antineoplastic Agents, Hormonal/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Clinical Trials as Topic
- Drug Evaluation, Preclinical
- Drug Resistance, Neoplasm
- Estrogen Antagonists/chemistry
- Estrogen Antagonists/pharmacology
- Estrogen Antagonists/therapeutic use
- Estrogen Receptor alpha/antagonists & inhibitors
- Estrogen Receptor alpha/chemistry
- Estrogen Receptor alpha/metabolism
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Models, Molecular
- Molecular Conformation
- Molecular Structure
- Mutation
- Protein Binding
- Protein Processing, Post-Translational
- Receptors, Estrogen/antagonists & inhibitors
- Receptors, Estrogen/chemistry
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Selective Estrogen Receptor Modulators/chemistry
- Selective Estrogen Receptor Modulators/pharmacology
- Selective Estrogen Receptor Modulators/therapeutic use
- Structure-Activity Relationship
- Treatment Outcome
Collapse
Affiliation(s)
- T Traboulsi
- Institute for Research in Immunology and CancerUniversité de Montréal, Montréal, Québec, Canada
- Department of Biochemistry and Molecular MedicineUniversité de Montréal, Montréal, Québec, Canada
| | - M El Ezzy
- Institute for Research in Immunology and CancerUniversité de Montréal, Montréal, Québec, Canada
| | - J L Gleason
- Department of ChemistryMcGill University, Montréal, Québec, Canada
| | - S Mader
- Institute for Research in Immunology and CancerUniversité de Montréal, Montréal, Québec, Canada
- Department of Biochemistry and Molecular MedicineUniversité de Montréal, Montréal, Québec, Canada
| |
Collapse
|
25
|
Ravikumara NR, Bharadwaj M, Madhusudhan B. Tamoxifen citrate-loaded poly(d,l) lactic acid nanoparticles: Evaluation for their anticancer activity in vitro and in vivo. J Biomater Appl 2016; 31:755-772. [PMID: 27664187 DOI: 10.1177/0885328216670561] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The optimization of tamoxifen citrate entrapment and its release from biodegradable poly(d,l) lactic acid nanoparticles are prepared by modified spontaneous emulsification solvent diffusion method. Since the addition of tamoxifen citrate induces the formation of drug crystals from nanoparticle suspension the influence of several parameters on tamoxifen citrate encapsulation was investigated. In vitro studies for cytotoxicity, DNA ladder, and the expression of Bcl-2-Bax expression were also investigated for MCF-7 and MDA-MB-231 cells after the addition of tamoxifen citrate alone and tamoxifen citrate-poly(d,l) lactic acid-nanoparticles (encapsulated tamoxifen citrate). From results, it was noticed that the size and zeta potential of the drug loaded nanoparticles were not differed much in their physicochemical properties from drug free counterparts. The drug-loaded and drug-free nanoparticles exhibited size of in between 271.4 and 282.7 nm and zeta potential of -34 to -27.4 mV, respectively. There was significant increase in drug incorporation in the particles noticed in dichloromethane + methanol system in comparison to acetone + methanol and ethyl acetate + methanol systems. The drug was partly released from the nanoparticles after 48 h of incubation at 37℃. From Fourier transform infrared spectroscopy and differential scanning calorimetry data demonstrated drug-polymer characteristics within the nanoparticles and unincorporated drug that appeared in the form of crystals from polarized microscopic study. MCF-7 and MDA-MB-231 cells were more sensitive to tamoxifen citrate-poly(d,l) lactic acid-nanoparticles than tamoxifen citrate alone. DNA ladder and the expression of Bax to Bcl-2 ratio were much higher in tamoxifen citrate encapsulated in nanoparticles than that in tamoxifen citrate alone. These results demonstrated the feasibility of encapsulation of tamoxifen citrate and its enhanced efficiency in vitro and in vivo studies.
Collapse
Affiliation(s)
- N R Ravikumara
- Department of Biochemistry, P.G. Centre, Research Center for Nanoscience and Technology, Kuvempu University, Shivagangotri, Davangere, India
| | - Mausumi Bharadwaj
- Institute of Cytology and Preventive Oncology Noida, Uttar Pradesh, India
| | | |
Collapse
|
26
|
Ho LA, Thomas E, McLaughlin RA, Flematti GR, Fuller RO. A new selective fluorescent probe based on tamoxifen. Bioorg Med Chem Lett 2016; 26:4879-4883. [PMID: 27662800 DOI: 10.1016/j.bmcl.2016.09.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 09/08/2016] [Accepted: 09/10/2016] [Indexed: 12/11/2022]
Abstract
Developing targeted validation probes that can interrogate biology is of interest for both chemists and biologists. The synthesis of suitable compounds provides a means for avoiding the costly labeling of cells with specific antibodies and the bias associated with the interpretation of biological validation experiments. The chemotherapeutic agent, tamoxifen has been routinely used in the treatment of breast cancer for decades. Once metabolized, the active form of tamoxifen (4-hydroxytamoxifen) competes with the binding of estrogens to the estrogen receptors (ER). Its selectivity in ER modulation makes it an ideal candidate for the development of materials to be used as chemical probes. Here we report the synthesis of a fluorescent BODIPY®FL conjugate of tamoxifen linked through an ethylene glycol moiety, and present proof-of-principle results in ER positive and ER negative cell lines. Optical microscopy indicates that the fluorescent probe binds selectively to tamoxifen sensitive breast cancer cell lines. The compound showed no affinity for the tamoxifen resistant breast cancer lines. The specificity of the new compound make it a valuable addition to the chemical probe tool kit for estrogen receptors.
Collapse
Affiliation(s)
- Louisa A Ho
- School of Chemistry and Biochemistry M310, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| | - Elizabeth Thomas
- School of Surgery M507, The University of Western Australia, QEII Medical Center, Monash Ave, Nedlands, WA 6009, Australia
| | - Robert A McLaughlin
- Australian Research Council Center of Excellence for Nanoscale Biophotonics, School of Medicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Gavin R Flematti
- School of Chemistry and Biochemistry M310, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia
| | - Rebecca O Fuller
- School of Chemistry and Biochemistry M310, The University of Western Australia, 35 Stirling Hwy, Crawley, WA 6009, Australia.
| |
Collapse
|
27
|
Ghosal P, Sukocheva OA, Wang T, Mayne GC, Watson DI, Hussey DJ. Effects of chemotherapy agents on Sphingosine-1-Phosphate receptors expression in MCF-7 mammary cancer cells. Biomed Pharmacother 2016; 81:218-224. [PMID: 27261597 DOI: 10.1016/j.biopha.2016.04.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 04/06/2016] [Accepted: 04/08/2016] [Indexed: 02/05/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a potent bioactive sphingolipid involved in the regulation of cell proliferation and cancer progression. Increased expression of S1P receptors has been detected in advanced breast tumours with poor prognosis suggesting that S1P receptors might control tumour response to chemotherapy. However, it remains unclear how the levels of S1P receptor expression are influenced by chemotherapy agents. Western immunoblotting, PCR analysis and fluorescent microscopy techniques were used in this study to analyze expression patterns of S1P receptors 2 and 3 (S1P2/S1P3) in MCF-7 breast adenocarcinoma cells treated by Tamoxifen (TAM) and/or Medroxyprogesterone acetate (MPA). We found that TAM/MPA induce downregulation of S1P3 receptors, but stimulate expression of S1P2. According to cell viability and caspase activity analyses, as expected, TAM activated apoptosis. We also detected TAM/MPA-induced autophagy marked by formation of macroautophagosomes and increased level of Beclin 1. Combined application of TAM and MPA resulted in synergistic apoptosis- and autophagy-stimulating effects. Assessed by fluorescent microscopy with autophagosome marker LAMP-2, changes in S1P receptor expression coincided with activation of autophagy, suggestively, directing breast cancer cells towards death. Further studies are warranted to explore the utility of manipulation of S1P2 and S1P3 receptor expression as a novel treatment approach.
Collapse
Affiliation(s)
- P Ghosal
- Flinders University Department of Surgery & Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Bedford Park, 5042, South Australia, Australia
| | - O A Sukocheva
- Flinders University Department of Surgery & Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Bedford Park, 5042, South Australia, Australia.
| | - T Wang
- Flinders University Department of Surgery & Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Bedford Park, 5042, South Australia, Australia
| | - G C Mayne
- Flinders University Department of Surgery & Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Bedford Park, 5042, South Australia, Australia
| | - D I Watson
- Flinders University Department of Surgery & Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Bedford Park, 5042, South Australia, Australia
| | - D J Hussey
- Flinders University Department of Surgery & Flinders Centre for Innovation in Cancer, Flinders Medical Centre, Bedford Park, 5042, South Australia, Australia
| |
Collapse
|
28
|
Swetzig WM, Wang J, Das GM. Estrogen receptor alpha (ERα/ESR1) mediates the p53-independent overexpression of MDM4/MDMX and MDM2 in human breast cancer. Oncotarget 2016; 7:16049-69. [PMID: 26909605 PMCID: PMC4941297 DOI: 10.18632/oncotarget.7533] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 01/27/2016] [Indexed: 12/31/2022] Open
Abstract
MDM2 and MDM4 are heterodimeric, non-redundant oncoproteins that potently inhibit the p53 tumor suppressor protein. MDM2 and MDM4 also enhance the tumorigenicity of breast cancer cells in in vitro and in vivo models and are overexpressed in primary human breast cancers. Prior studies have characterized Estrogen Receptor Alpha (ERα/ESR1) as a regulator of MDM2 expression and an MDM2- and p53-interacting protein. However, similar crosstalk between ERα and MDM4 has not been investigated. Moreover, signaling pathways that mediate the overexpression of MDM4 in human breast cancer remain to be elucidated. Using the Cancer Genome Atlas (TCGA) breast invasive carcinoma patient cohort, we have analyzed correlations between ERα status and MDM4 and MDM2 expression in primary, treatment-naïve, invasive breast carcinoma samples. We report that the expression of MDM4 and MDM2 is elevated in primary human breast cancers of luminal A/B subtypes and associates with ERα-positive disease, independently of p53 mutation status. Furthermore, in cell culture models, ERα positively regulates MDM4 and MDM2 expression via p53-independent mechanisms, and these effects can be blocked by the clinically-relevant endocrine therapies fulvestrant and tamoxifen. Additionally, ERα also positively regulates p53 expression. Lastly, we report that endogenous MDM4 negatively regulates ERα expression and forms a protein complex with ERα in breast cancer cell lines and primary human breast tumor tissue. This suggests direct signaling crosstalk and negative feedback loops between ERα and MDM4 expression in breast cancer cells. Collectively, these novel findings implicate ERα as a central component of the p53-MDM2-MDM4 signaling axis in human breast cancer.
Collapse
Affiliation(s)
- Wendy M. Swetzig
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Molecular Pharmacology and Cancer Therapeutics, The University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Jianmin Wang
- Department of Bioinformatics and Biostatistics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Gokul M. Das
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY, USA
- Department of Molecular Pharmacology and Cancer Therapeutics, The University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
29
|
Claro de Souza M, Marotta-Oliveira SS, Rocha NHS, Eloy JO, Marchetti JM. Development of a Method to Evaluate the Release Profile of Tamoxifen from Pegylated Hybrid Micelles. J LIQ CHROMATOGR R T 2015. [DOI: 10.1080/10826076.2015.1032418] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Marina Claro de Souza
- College of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paolo, Brazil
| | | | - Nathália Helena Souza Rocha
- College of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paolo, Brazil
| | - Josimar O. Eloy
- College of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paolo, Brazil
| | - Juliana Maldonado Marchetti
- College of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paolo, Brazil
| |
Collapse
|
30
|
Abstract
BACKGROUND It appears that the female reproductive events and hormonal treatments may impact the course of bipolar disorder in women. In particular, childbirth is known to be associated with onset of affective episodes in women with bipolar disorder. During the female reproductive events the sex hormones, e.g. estrogen, are fluctuating and particularly postpartum there is a steep fall in the levels of serum estrogen. The role of estrogen in women with bipolar disorder is, however, not fully understood. AIM The main objective of this review is to evaluate the possible relation between serum estrogen levels and women with bipolar disorder including studies of the anti manic effects of the selective estrogen receptor modulator tamoxifen. METHOD A systematically literature search on PubMed was conducted: two studies regarding the connection between serum estrogen levels and women with bipolar disorder were identified. Furthermore, four studies were found concerning the antimanic effects of tamoxifen. RESULTS Both studies in the estrogen studies showed very low levels of estrogen in women with postpartum psychosis and significant improvement of symptoms after treatment with estrogen. The four tamoxifen studies found that tamoxifen was effective in producing antimanic effects. CONCLUSION These results indicate that estrogen fluctuations may be an important factor in the etiology of bipolar disorder and it is obvious that more research on this topic is needed to clarify the role of estrogen in women with bipolar disorder.
Collapse
Affiliation(s)
- Ninja Meinhard
- Psychiatric Center Copenhagen, Rigshospitalet, University Hospital of Copenhagen , Blegdamsvej 9, DK-2100 Copenhagen , Denmark
| | | | | |
Collapse
|
31
|
Güngör S, Delgado-Charro MB, Masini-Etévé V, Potts RO, Guy RH. Transdermal flux predictions for selected selective oestrogen receptor modulators (SERMs): Comparison with experimental results. J Control Release 2013; 172:601-6. [DOI: 10.1016/j.jconrel.2013.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 09/03/2013] [Accepted: 09/19/2013] [Indexed: 11/28/2022]
|
32
|
Visser K, Mortimer M, Louw A. Cyclopia extracts act as ERα antagonists and ERβ agonists, in vitro and in vivo. PLoS One 2013; 8:e79223. [PMID: 24223909 PMCID: PMC3817056 DOI: 10.1371/journal.pone.0079223] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/20/2013] [Indexed: 12/28/2022] Open
Abstract
Hormone replacement therapy associated risks, and the concomitant reluctance of usage, has instigated the search for new generations of estrogen analogues that would maintain estrogen benefits without associated risks. Furthermore, if these analogues display chemo-preventative properties in breast and endometrial tissues it would be of great value. Both the selective estrogen receptor modulators as well as the selective estrogen receptor subtype modulators have been proposed as estrogen analogues with improved risk profiles. Phytoestrogen containing extracts of Cyclopia, an indigenous South African fynbos plant used to prepare Honeybush tea may serve as a source of new estrogen analogues. In this study three extracts, P104, SM6Met, and cup-of-tea, from two species of Cyclopia, C. genistoides and C. subternata, were evaluated for ER subtype specific agonism and antagonism both in transactivation and transrepression. For transactivation, the Cyclopia extracts displayed ERα antagonism and ERβ agonism when ER subtypes were expressed separately, however, when co-expressed only agonism was uniformly observed. In contrast, for transrepression, this uniform behavior was lost, with some extracts (P104) displaying uniform agonism, while others (SM6Met) displayed antagonism when subtypes were expressed separately and agonism when co-expressed. In addition, breast cancer cell proliferation assays indicate that extracts antagonize cell proliferation in the presence of estrogen at lower concentrations than that required for proliferation. Furthermore, lack of uterine growth and delayed vaginal opening in an immature rat uterotrophic model validates the ERα antagonism of extracts observed in vitro and supports the potential of the Cyclopia extracts as a source of estrogen analogues with a reduced risk profile.
Collapse
Affiliation(s)
- Koch Visser
- Department of Biochemistry, University of Stellenbosch, Matieland, Stellenbosch, Republic of South Africa
| | - Morné Mortimer
- Department of Biochemistry, University of Stellenbosch, Matieland, Stellenbosch, Republic of South Africa
| | - Ann Louw
- Department of Biochemistry, University of Stellenbosch, Matieland, Stellenbosch, Republic of South Africa
- * E-mail:
| |
Collapse
|
33
|
Combined effects of goserelin and tamoxifen on estradiol level, breast density, and endometrial thickness in premenopausal and perimenopausal women with early-stage hormone receptor-positive breast cancer: a randomised controlled clinical trial. Br J Cancer 2013; 109:582-8. [PMID: 23860520 PMCID: PMC3738136 DOI: 10.1038/bjc.2013.324] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 05/28/2013] [Accepted: 06/04/2013] [Indexed: 12/02/2022] Open
Abstract
Background: This study is to investigate the effects of geserelin+tamoxifen (TAM) on estradiol level, breast density (BD), endometrial thickness (ET), and blood lipids in premenopausal and perimenopausal women with hormone receptor-positive early-stage breast cancer. Methods: This study recruited 110 premenopausal and perimenopausal patients with hormone receptor-positive early-stage breast cancer between 22 June 2008 and 31 December 2009 and randomly assigned them to receive either goserelin plus TAM or TAM alone for 1.5 years. Blood levels of sex hormones and lipids and ET were determined at 0, 3, 6, 12, and 18 months. Contralateral BD was also measured at 0, 12, and 18 months. Results: Five participants dropped out of the goserelin plus TAM group, and two participants dropped out of the TAM-alone group before initiation of endocrine therapy. The rest of patients received scheduled treatment and 3 years of median follow-up. No serious adverse effects were observed, and only two local recurrences have been observed in these patients. Estradiol level and BD were lower in the goserelin plus TAM group than in the TAM-alone group (P<0.05). The endometrium in the goserelin plus TAM group was significantly thinner than that in the TAM-alone group (P<0.05), and women in the TAM-alone group exhibited endometrial thickening over the course of the study. Furthermore, no significant differences in blood lipid levels were reported between the two groups. Conclusion: The data from the current study demonstrated that the addition of goserelin to TAM results in downregulation of estradiol level, followed by significant reduction in BD and ET in premenopausal and perimenopausal women with hormone receptor-positive breast cancer, which may eventually lead to better outcome in these patients.
Collapse
|
34
|
|
35
|
Dragnev K, You M, Wang Y, Lubet R. Lung cancer chemoprevention: difficulties, promise and potential agents? Expert Opin Investig Drugs 2012; 22:35-47. [PMID: 23167766 DOI: 10.1517/13543784.2013.731392] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION In a variety of cancers there is evidence that specific regimens can prevent or significantly delay the development of cancer. Thus, for breast cancer (ER+) use of SERMs or aromatase inhibitors can substantially decrease tumor incidence. For cervical cancer, HPV vaccination will inhibit long term cancer incidence. For colon cancer, the second greatest cancer killer, administration of aspirin and other NSAIDs decreases advanced colon adenomas in Phase II trials and epidemiologic data support their ability to prevent colon cancer. To date prevention trials in the area of lung cancer have shown minimal efficacy. AREAS COVERED The paper examines and discusses in greater detail certain promising agents which the authors have tested either preclinically and or in early phase clinical trials. These agents include RXR agonists, EGFr inhibitors, NSAIDs and Triterpenoids. Other agents including glucocorticoids, pioglitazone and iloprost are briefly mentioned. In addition, the paper presents various types of potential Phase II lung cancer prevention trials and describes their strengths and weaknesses. The potential use of various biomarkers as endpoints in trials e.g. histopathology, non-specific biomarkers (e.g., Ki67, cyclin D expression, apoptosis) and molecular biomarkers (e.g. specific phosphorylated proteins, gene expression etc.) is presented. Finally, we examine at least one approach, the use of aerosols, which may diminish the systemic toxicity associated with certain of these agents. EXPERT OPINION The manuscript presents: a) a number of promising agents which appear applicable to further Phase II prevention trials; b) approaches to defining potential preventive agents as well; c) approaches which might mitigate the side effects associated with potential agents most specifically the use of aerosols. Finally, we discuss biomarker studies both preclinical and clinical which might help support potential Phase II trials. The particular appeal to the preclinical studies is that they can be followed to a tumor endpoint. We hope that this will give the reader further background and allow one to appreciate the potential and some of the hurdles associated with lung cancer chemoprevention.
Collapse
|
36
|
Bhatia A, Singh B, Raza K, Shukla A, Amarji B, Katare OP. Tamoxifen-loaded novel liposomal formulations: evaluation of anticancer activity on DMBA-TPA induced mouse skin carcinogenesis. J Drug Target 2012; 20:544-50. [DOI: 10.3109/1061186x.2012.694887] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Blanco MD, Benito M, Olmo R, Teijón C, Pérez E, Katime I, Teijón JM. Synthesis and in vitro biological evaluation as antitumour drug carriers of folate-targeted N-isopropylacrylamide-based nanohydrogels. POLYM INT 2012. [DOI: 10.1002/pi.4200] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
38
|
Ellis GA, McGrath NA, Palte MJ, Raines RT. Ribonuclease-Activated Cancer Prodrug. ACS Med Chem Lett 2012; 3:268-272. [PMID: 22611478 DOI: 10.1021/ml2002554] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Cancer chemotherapeutic agents often have a narrow therapeutic index that challenges the maintenance of a safe and effective dose. Consistent plasma concentrations of a drug can be obtained by using a timed-release prodrug strategy. We reasoned that a ribonucleoside 3'-phosphate could serve as a pro-moiety that also increases the hydrophilicity of a cancer chemotherapeutic agent. Herein, we report an efficient route for the synthesis of the prodrug uridine 3'-(4-hydroxytamoxifen phosphate) (UpHT). UpHT demonstrates timed-released activation kinetics with a half-life of approximately 4 h at the approximate plasma concentration of human pancreatic ribonuclease (RNase 1). MCF-7 breast cancer cells treated with UpHT showed decreased proliferation upon co-incubation with RNase 1, consistent with the release of the active drug-4-hydroxytamoxifen. These data demonstrate the utility of a human plasma enzyme as a useful activator of a prodrug.
Collapse
Affiliation(s)
- Gregory A. Ellis
- Department of Biochemistry, ‡Department of Chemistry, and §Medical Scientist Training Program and Molecular & Cellular Pharmacology Graduate Training Program, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Nicholas A. McGrath
- Department of Biochemistry, ‡Department of Chemistry, and §Medical Scientist Training Program and Molecular & Cellular Pharmacology Graduate Training Program, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Michael J. Palte
- Department of Biochemistry, ‡Department of Chemistry, and §Medical Scientist Training Program and Molecular & Cellular Pharmacology Graduate Training Program, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Ronald T. Raines
- Department of Biochemistry, ‡Department of Chemistry, and §Medical Scientist Training Program and Molecular & Cellular Pharmacology Graduate Training Program, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
39
|
Fenández A, Teijón C, Benito M, Iglesias I, Lozano R, Teijón JM, Blanco MD. Tamoxifen-loaded microspheres based on mixtures of poly(D,L-lactide-co-glycolide) and poly(D,L-lactide) polymers: Effect of polymeric composition on drug release and in vitro antitumoral activity. J Appl Polym Sci 2011. [DOI: 10.1002/app.35327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
40
|
Sarmah JK, Mahanta R, Bhattacharjee SK, Mahanta R, Biswas A. Controlled release of tamoxifen citrate encapsulated in cross-linked guar gum nanoparticles. Int J Biol Macromol 2011; 49:390-6. [PMID: 21641924 DOI: 10.1016/j.ijbiomac.2011.05.020] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2011] [Revised: 05/19/2011] [Accepted: 05/19/2011] [Indexed: 10/18/2022]
Abstract
Natural polysaccharides, due to their outstanding merits, have received more and more attention in the field of drug delivery. In the present study tamoxifen citrate, TMX (a non-steroidal antiestrogenic drug) loaded guar gum nanoparticles, GG NPs, crosslinked with glutaraldehyde were prepared for treatment of breast cancer. An oil in water (o/w) emulsion polymer cross-linking method was employed for preparation of blank and drug loaded sustained release nature biodegradable nanoparticles. Prepared nanoparticles were characterized by morphology in scanning electron microscope (SEM), size distribution in transmission electron microscope (TEM), TMX loading by high performance liquid chromatography (HPLC) and in vitro drug release characteristics. An overall sustained release of the drug from the biodegradable nanoparticles was observed in in vitro release studies. The release of TMX from GG NPs was found to be effected by guar gum and glutaraldehyde concentration. Regression coefficient (R(2)) analysis suggested that the predominant mechanism behind the drug release from the nanoparticles was time dependent release and diffusion. In vivo studies on female albino mice demonstrated maximum uptake of the drug by mammary tissue after 24h of administration with drug loaded guar gum nanoparticles in comparison with that with the tablet form of the drug. These findings demonstrate that controlled release of TMX from GG NPs could be a potential alternative pharmaceutical formulation in passive targeting of TMX in breast cancer treatments.
Collapse
|
41
|
Sarzehi S, Chamani J. Investigation on the interaction between tamoxifen and human holo-transferrin: Determination of the binding mechanism by fluorescence quenching, resonance light scattering and circular dichroism methods. Int J Biol Macromol 2010; 47:558-69. [DOI: 10.1016/j.ijbiomac.2010.08.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 08/01/2010] [Accepted: 08/04/2010] [Indexed: 01/26/2023]
|
42
|
Blanco MD, Guerrero S, Benito M, Teijón C, Olmo R, Muñíz E, Katime I, Teijón JM. Tamoxifen-loaded folate-conjugate poly[(p-nitrophenyl acrylate)-co-(N-isopropylacrylamide)] sub-microgel as antitumoral drug delivery system. J Biomed Mater Res A 2010; 95:1028-40. [DOI: 10.1002/jbm.a.32929] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 05/12/2010] [Accepted: 07/01/2010] [Indexed: 11/07/2022]
|
43
|
Sahana B, Santra K, Basu S, Mukherjee B. Development of biodegradable polymer based tamoxifen citrate loaded nanoparticles and effect of some manufacturing process parameters on them: a physicochemical and in-vitro evaluation. Int J Nanomedicine 2010; 5:621-30. [PMID: 20856837 PMCID: PMC2939707 DOI: 10.2147/ijn.s9962] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The aim of the present study was to develop nanoparticles of tamoxifen citrate, a non-steroidal antiestrogenic drug used for the treatment of breast cancer. Biodegradable poly (D, L- lactide-co-glycolide)-85:15 (PLGA) was used to develop nanoparticles of tamoxifen citrate by multiple emulsification (w/o/w) and solvent evaporation technique. Drug-polymer ratio, polyvinyl alcohol concentrations, and homogenizing speeds were varied at different stages of preparation to optimize the desired size and release profile of drug. The characterization of particle morphology and shape was performed by field emission scanning electron microscope (FE-SEM) and particle size distribution patterns were studied by direct light scattering method using zeta sizer. In vitro drug release study showed that release profile of tamoxifen from biodegradable nanoparticles varied due to the change in speed of centrifugation for separation. Drug loading efficiency varied from 18.60% to 71.98%. The FE-SEM study showed that biodegradable nanoparticles were smooth and spherical in shape. The stability studies of tamoxifen citrate in the experimental nanoparticles showed the structural integrity of tamoxifen citrate in PLGA nanoparticles up to 60°C in the tested temperatures. Nanoparticles containing tamoxifen citrate could be useful for the controlled delivery of the drug for a prolonged period.
Collapse
Affiliation(s)
- Basudev Sahana
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | | | | | | |
Collapse
|
44
|
Layek B, Mukherjee B. Tamoxifen citrate encapsulated sustained release liposomes: preparation and evaluation of physicochemical properties. Sci Pharm 2010; 78:507-15. [PMID: 21179362 PMCID: PMC3002818 DOI: 10.3797/scipharm.0911-11] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2009] [Accepted: 07/12/2010] [Indexed: 12/02/2022] Open
Abstract
The present study was designed for the development of a stable sustained release liposomal drug delivery system for tamoxifen citrate (TC) using soya phosphatidylcholine (SPC), cholesterol (CH) and span 20 as main ingredients. Liposomes were prepared by formation of thin lipid film followed by hydration. The mean vesicle diameter was found to be 203.5 ± 19.5 nm with 21% of the liposomal population having average diameter below 76.72 ± 6.7 nm. There was a good vesicular distribution with the polydispersity index of 0.442 ± 0.03. The maximum loading of drug was determined to be 53.60% of the initial amount that is 34.58 μg of drug per mg of lipid. Amongst the different storage conditions, liposomes stored at 2–8°C were found to be most stable and only 4% of the drug was lost over the storage period of 5 weeks. In vitro release studies of liposomes showed that 50% of drug was released within 3 hours (h) whereas 95% drug was released in 30 h. This indicates the usefulness of the liposomal delivery system for sustaining the in vitro release of tamoxifen citrate.
Collapse
Affiliation(s)
- Buddhadev Layek
- Department of Pharmaceutical Technology, Division of Pharmaceutics, Jadavpur University, Kolkata-700032, India.
| | | |
Collapse
|
45
|
Kieser KJ, Kim DW, Carlson KE, Katzenellenbogen BS, Katzenellenbogen JA. Characterization of the pharmacophore properties of novel selective estrogen receptor downregulators (SERDs). J Med Chem 2010; 53:3320-9. [PMID: 20334372 DOI: 10.1021/jm100047k] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Selective estrogen receptor (ER) down-regulators (SERDs) reduce ERalpha protein levels as well as block ER activity and therefore are promising therapeutic agents for the treatment of hormone refractory breast cancer. Starting with the triarylethylene acrylic acid SERD 4, we have investigated how alterations in both the ligand core structure and the appended acrylic acid substituent affect SERD activity. The new ligands were based on high affinity, symmetrical cyclofenil or bicyclo[3.3.1]nonane core systems, and in these, the position of the carboxyl group was extended from the ligand core, either retaining the vinylic linkage of the substituent or replacing it with an ether linkage. Although most structural variants showed binding affinities for ERalpha and ERbeta higher than that of 4, only the compounds preserving the acrylic acid side chain retained SERD activity, although they could possess varying core structures. Hence, the acrylic acid moiety of the ligand is crucial for SERD-like blockade of ER activities.
Collapse
Affiliation(s)
- Karen J Kieser
- Department of Chemistry, University of Illinois, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
| | | | | | | | | |
Collapse
|
46
|
Kok M, Zwart W, Holm C, Fles R, Hauptmann M, Van't Veer LJ, Wessels LFA, Neefjes J, Stål O, Linn SC, Landberg G, Michalides R. PKA-induced phosphorylation of ERα at serine 305 and high PAK1 levels is associated with sensitivity to tamoxifen in ER-positive breast cancer. Breast Cancer Res Treat 2010; 125:1-12. [PMID: 20213082 DOI: 10.1007/s10549-010-0798-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Accepted: 02/09/2010] [Indexed: 01/30/2023]
Abstract
Phosphorylation of estrogen receptor α at serine 305 (ERαS305-P) by protein kinase A (PKA) or p21-activated kinase 1 (PAK1) has experimentally been associated with tamoxifen sensitivity. Here, we investigated the clinical application of this knowledge to predict tamoxifen resistance in ER-positive breast cancer patients. Using immunohistochemistry, a score including PAK1 and co-expression of PKA and ERαS305-P (PKA/ERαS305-P) was developed on a training set consisting of 103 patients treated with tamoxifen for metastatic disease, and validated on 231 patients randomized between adjuvant tamoxifen or no treatment. In the training set, PAK1 levels were associated with tumor progression after tamoxifen (HR 1.57, 95% CI 0.99-2.48), as was co-expression of PKA and ERαS305-P (HR 2.00, 95% CI 1.14-3.52). In the validation set, a significant tamoxifen benefit was found among the 73% patients negative for PAK1 and PKA/ERαS305-P (HR 0.54, 95% CI 0.34-0.87), while others (27%) were likely to have no benefit from tamoxifen (HR 0.88, 95% 0.42-1.82). The test for interaction showed a significant difference in recurrence-free survival between groups defined by PAK1 and PKA/ERαS305-P (P = 0.037). Elevated PAK1 and PKA/ERαS305-P appeared to influence tamoxifen sensitivity. Both PAK1 and PKA/ERαS305-P levels were associated with sensitivity to tamoxifen in breast tumors and the combination of these variables should be considered in predicting tamoxifen benefit.
Collapse
Affiliation(s)
- Marleen Kok
- Department of Experimental Therapy, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Preparation of cross-linked guar gum nanospheres containing tamoxifen citrate by single step emulsion in situ polymer cross-linking method. J INCL PHENOM MACRO 2009. [DOI: 10.1007/s10847-009-9589-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
48
|
Ball LJ, Levy N, Zhao X, Griffin C, Tagliaferri M, Cohen I, Ricke WA, Speed TP, Firestone GL, Leitman DC. Cell type- and estrogen receptor-subtype specific regulation of selective estrogen receptor modulator regulatory elements. Mol Cell Endocrinol 2009; 299:204-11. [PMID: 19059307 PMCID: PMC3420066 DOI: 10.1016/j.mce.2008.10.050] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Revised: 10/08/2008] [Accepted: 10/24/2008] [Indexed: 11/28/2022]
Abstract
Selective estrogen receptor modulators (SERMs), such as tamoxifen and raloxifene can act as estrogen receptor (ER) antagonists or agonists depending on the cell type. The antagonistic action of tamoxifen has been invaluable for treating breast cancer, whereas the agonist activity of SERMs also has important clinical applications as demonstrated by the use of raloxifene for osteoporosis. Whereas the mechanism whereby SERMs function as antagonists has been studied extensively very little is known about how SERMs produce agonist effects in different tissues with the two ER types; ERalpha and ERbeta. We examined the regulation of 32 SERM-responsive regions with ERalpha and ERbeta in transiently transfected MCF-7 breast, Ishikawa endometrial, HeLa cervical and WAR-5 prostate cancer cells. The regions were regulated by tamoxifen and raloxifene in some cell types, but not in all cell lines. Tamoxifen activated similar number of regions with ERalpha and ERbeta in the cell lines, whereas raloxifene activated over twice as many regions with ERbeta compared to ERalpha. In Ishikawa endometrial cancer cells, tamoxifen activated 17 regions with ERalpha, whereas raloxifene activated only 2 regions, which might explain their different effects on the endometrium. Microarray studies also found that raloxifene regulated fewer genes than tamoxifen in U2OS bone cancer cells expressing ERalpha, whereas tamoxifen was equally effective at regulating genes with ERalpha and ERbeta. Our studies indicate that tamoxifen is a non-selective agonist, whereas raloxifene is a relative ERbeta-selective agonist, and suggest that ERbeta-selective SERMs might be safer for treating clinical conditions that are dependent on the agonist property of SERMs.
Collapse
Affiliation(s)
- Lonnele J. Ball
- Departments of Obstetrics, Gynecology and Reproductive Sciences, Cellular and Molecular Pharmacology, Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | - Nitzan Levy
- Departments of Obstetrics, Gynecology and Reproductive Sciences, Cellular and Molecular Pharmacology, Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | | | - Chandi Griffin
- Departments of Obstetrics, Gynecology and Reproductive Sciences, Cellular and Molecular Pharmacology, Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
| | | | | | - William A. Ricke
- Department of Urology, James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Terence P. Speed
- Department of Statistics, University of California, Berkeley, CA 94720, USA
- Division of Bioinformatics, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Gary L. Firestone
- Department of Molecular and Cell Biology and The Cancer Research Laboratory, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Dale C. Leitman
- Departments of Obstetrics, Gynecology and Reproductive Sciences, Cellular and Molecular Pharmacology, Center for Reproductive Sciences, University of California, San Francisco, CA 94143, USA
- Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, CA 94720, USA
- Corresponding author at: Departments of Obstetrics, Gynecology and Reproductive Sciences, Cellular and Molecular Pharmacology, University of California, San Francisco, S1258 P.O. Box 0556, San Francisco, CA 94143-0556, USA. Tel.: +1 415 502 5262. (D.C. Leitman)
| |
Collapse
|
49
|
Licciardi M, Craparo EF, Giammona G, Armes SP, Tang Y, Lewis AL. in vitro biological evaluation of folate-functionalized block copolymer micelles for selective anti-cancer drug delivery. Macromol Biosci 2008; 8:615-26. [PMID: 18432597 DOI: 10.1002/mabi.200800009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The main objective of this study was to evaluate the ability of folic acid-functionalized diblock copolymer micelles to improve the delivery and uptake of two poorly water-soluble anti-tumor drugs, tamoxifen and paclitaxel, to cancer cells through folate receptor targeting. The diblock copolymer used in this study comprised a hydrophilic poly[2-(methacryloyloxy)ethyl phosphorylcholine] (MPC) block, carrying at the chain end the folate targeting moiety, and a pH-sensitive hydrophobic poly[2-(diisopropylamino)ethyl methacrylate] (DPA) block (FA-MPC-DPA). The drug-loading capacities of tamoxifen- and paclitaxel-loaded micelles were determined by high performance liquid chromatography and the micelle dimensions were determined by dynamic light scattering and transmission electron microscopy. Cell viability studies were carried out on human chronic myelogenous leukaemia (K-562) and colon carcinoma cell lines (Caco-2) in order to demonstrate that drug-loaded FA-MPC-DPA micelles exhibited higher cytotoxicities toward cancer cells than unfunctionalized MPC-DPA micelles. Uptake studies confirmed that folate-conjugated micelles led to increased drug uptake within cancer cells, demonstrating the expected selectivity toward these tumor cells.
Collapse
Affiliation(s)
- Mariano Licciardi
- Dipartimento di Chimica e Tecnologie Farmaceutiche, Via Archirafi 32, 90123 Palermo, Italy
| | | | | | | | | | | |
Collapse
|
50
|
Nishihori T, Choi J, DiGiovanna MP, Thomson JG, Kohler PC, McGurn J, Chung GG. Carpal tunnel syndrome associated with the use of aromatase inhibitors in breast cancer. Clin Breast Cancer 2008; 8:362-5. [PMID: 18757265 DOI: 10.3816/cbc.2008.n.043] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Aromatase inhibitors (AI) inhibit peripheral conversion of androgens to estradiol and are commonly used as hormonal therapy for postmenopausal women with hormone receptor-positive breast cancer in the metastatic and adjuvant settings. Joint-related symptoms, however, are seen in a significant proportion of patients. Carpal tunnel syndrome (CTS) is a common nerve entrapment disorder affecting the median nerve. We describe 6 patients with newly diagnosed CTS after initiation of adjuvant AI therapy. Aromatase inhibitors were discontinued in several patients secondary to this toxicity with some switching to tamoxifen and most subsequently experiencing relief of their symptoms. Potential pathophysiologic roles of hormonal manipulation with AIs and other risk factors that might contribute to CTS are discussed. Aromatase inhibitors might accentuate the occurrence of CTS and potentially other nerve entrapment syndromes, and a more systematic approach should be used to better understand the clinical significance and incidence of these symptoms.
Collapse
Affiliation(s)
- Taiga Nishihori
- Department of Internal Medicine, Medical Oncology, Yale University School of Medicine, Yale Comprehensive Cancer Center, New Haven, CT, USA
| | | | | | | | | | | | | |
Collapse
|