1
|
Kofman K, Levin M. Bioelectric pharmacology of cancer: A systematic review of ion channel drugs affecting the cancer phenotype. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 191:25-39. [PMID: 38971325 DOI: 10.1016/j.pbiomolbio.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/21/2024] [Accepted: 07/04/2024] [Indexed: 07/08/2024]
Abstract
Cancer is a pernicious and pressing medical problem; moreover, it is a failure of multicellular morphogenesis that sheds much light on evolutionary developmental biology. Numerous classes of pharmacological agents have been considered as cancer therapeutics and evaluated as potential carcinogenic agents; however, these are spread throughout the primary literature. Here, we briefly review recent work on ion channel drugs as promising anti-cancer treatments and present a systematic review of the known cancer-relevant effects of 109 drugs targeting ion channels. The roles of ion channels in cancer are consistent with the importance of bioelectrical parameters in cell regulation and with the functions of bioelectric signaling in morphogenetic signals that act as cancer suppressors. We find that compounds that are well-known for having targets in the nervous system, such as voltage-gated ion channels, ligand-gated ion channels, proton pumps, and gap junctions are especially relevant to cancer. Our review suggests further opportunities for the repurposing of numerous promising candidates in the field of cancer electroceuticals.
Collapse
Affiliation(s)
- Karina Kofman
- Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Michael Levin
- Allen Discovery Center at Tufts University, USA; Wyss Institute for Biologically Inspired Engineering at Harvard University, USA.
| |
Collapse
|
2
|
Aziz N, Faraz M, Sherwani MA, Fatma T, Prasad R. Illuminating the Anticancerous Efficacy of a New Fungal Chassis for Silver Nanoparticle Synthesis. Front Chem 2019; 7:65. [PMID: 30800654 PMCID: PMC6375905 DOI: 10.3389/fchem.2019.00065] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/23/2019] [Indexed: 01/16/2023] Open
Abstract
Biogenic silver nanoparticles (Ag NPs) have supple platforms designed for biomedical and therapeutic intervention. Utilization of Ag NPs are preferred in the field of biomedicines and material science research because of their antioxidant, antimicrobial, and anticancerous activity along with their eco-friendly, biocompatible, and cost-effective nature. Here we present a novel fungus Piriformospora indica as an excellent source for obtaining facile and reliable Ag NPs with a high degree of consistent morphology. We demonstrated their cytotoxic property, coupled with their intrinsic characteristic that make these biogenic nanoparticles suitable for the anticancerous activity. In vitro cytotoxicity of biologically synthesized Ag NPs (BSNPs) and chemically synthesized Ag NPs (SNPs) was screened on various cancer cell lines, such as Human breast adenocarcinoma (MCF-7), Human cervical carcinoma (HeLa), Human liver hepatocellular carcinoma (HepG2) cell lines and embryonic kidney cell line (HEK-293) as normal cell lines. The antiproliferative outcome revealed that the BSNPs exhibited significant cytotoxic activity against MCF-7 followed by HeLa and HepG2 cell lines as compared to SNPs. The blend of cytotoxic properties, together with green and cost-effective characteristics make up these biogenic nanoparticles for their potential applications in cancer nanomedicine and fabrication coating of ambulatory and non-ambulatory medical devices.
Collapse
Affiliation(s)
- Nafe Aziz
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Mohd Faraz
- Department of Physics, Indian Institute of Technology Delhi, Haus Khas, New Delhi, India
| | - Mohd Asif Sherwani
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, India
| | - Tasneem Fatma
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Ram Prasad
- School of Environmental Science and Engineering, Sun Yat-Sen University, Guangzhou, China.,Amity Institute of Microbial Technology, Amity University, Noida, India
| |
Collapse
|
3
|
Zanganeh N, Ziamajidi N, Khodadadi I, Saidijam M, Abbasalipourkabir R. Liver Genes Expression Induced by Tamoxifen Loaded Solid Lipid Nanoparticles in Wistar Female Rats. Cell Biochem Biophys 2017; 76:303-310. [PMID: 29090414 DOI: 10.1007/s12013-017-0833-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 10/12/2017] [Indexed: 11/27/2022]
Abstract
The objective of this study was to investigate the effect of free tamoxifen and tamoxifen-loaded solid lipid nanoparticles (SLN) on cytochrome P450 (CYP3A2) and flavin-containing monooxygenase1 (FMO1) genes expression in the liver of female Wistar rats. Thirty female Wistar rats aged 7-8 weeks, were divided into six groups of six rats each. The first, second, third, and fourth groups were ovariectomized and received tamoxifen (2 mg/kg of body weight dissolved in 1 ml olive oil), tamoxifen-loaded SLN (2 mg/kg of body weight dispersed in 1 ml olive oil), SLN (10 mg/kg of body weight dispersed in 1 ml olive oil), and 1 ml olive oil, respectively. The fifth group comprised untreated ovariectomized control group and the sixth group served as unovariectomized healthy group. The treatments were given orally to the animals on 21 consecutive days using gastric intubations. At the end of the study, the rats were scarified and studied for some serum biochemical profile and two liver genes expression. The group treated with tamoxifen-loaded SLN showed significantly increased gene expression of CYP3A2 in comparison with the control, healthy, and group treated with free tamoxifen. The gene expression of FMO1 in the group that received tamoxifen-loaded SLN was significantly lower than that in the group treated with free tamoxifen. In addition, the group treated with free tamoxifen showed significantly increased gene expression of FMO1 as compared to the control and healthy groups. Encapsulation of tamoxifen inside solid lipid nanoparticles increased the gene expression of CYP3A2 and decreased the gene expression of FMO1.
Collapse
Affiliation(s)
- Naser Zanganeh
- Department of Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nasrin Ziamajidi
- Department of Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Khodadadi
- Department of Biochemistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Massoud Saidijam
- Department of Medical Biotechnology, Hamadan University of Medical Sciences, Hamadan, Iran
| | | |
Collapse
|
4
|
Ezzat A, Abdelhamid AO, El Awady MK, Abd El Azeem AS, Mohammed DM. The biochemical effects of nano tamoxifen and some bioactive components in experimental breast cancer. Biomed Pharmacother 2017; 95:571-576. [PMID: 28869895 DOI: 10.1016/j.biopha.2017.08.099] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 08/11/2017] [Accepted: 08/23/2017] [Indexed: 01/06/2023] Open
Abstract
The effect of nano tamoxifen and some bioactive components such as yeast, isoflavone, and silymarin on the level of resistance and prevention of breast cancer progression in experimental animals is the target of this study. Thirty female Sprague-Dawley rats received a single medication dosage of 7,12-dimethylbenz[a]anthracene (DMBA) intragastrically. After fourteen days of DMBA admission, the procedure protocol started out. Finally, all the experimental results evaluated, tabulated and statistically analyzed. The results demonstrated a highly significant elevation in the 8-OHdG level in group 1 (nano yeast) and 3 (nano silymarin) while the results demonstrated a highly significant reduction in group 2 (nano tamoxifen). The apoptosis results demonstrated a significant elevation in group 3 (nano silymarin) where appeared significant reduction in group 4 (nano isoflavone). ErbB-2 results demonstrated a significant elevation in group 2 (nano tamoxifen) and a significant reduction in each of group 3 (nano silymarin) and 4 (nano isoflavone). The lipid peroxide level demonstrated an extremely significant reduction in group 4 (nano isoflavone). And a significant reduction of total antioxidant was observed in group 3 (nano silymarin) in comparison to injected animals control. This may be considered a new vision and strategy to resist breast cancer disease or prevent progression.
Collapse
Affiliation(s)
- Afaf Ezzat
- Department of Nutrition and Food Science, National Research Centre, Dokki, Cairo 12622, Egypt
| | | | - Mostafa K El Awady
- Microbial Biotechnology Department, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Amal S Abd El Azeem
- Department of Nutrition and Food Science, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Dina Mostafa Mohammed
- Department of Nutrition and Food Science, National Research Centre, Dokki, Cairo 12622, Egypt.
| |
Collapse
|
5
|
El-Shorbagy HM. Potential anti-genotoxic effect of sodium butyrate to modulate induction of DNA damage by tamoxifen citrate in rat bone marrow cells. Cytotechnology 2017; 69:89-102. [PMID: 27905024 PMCID: PMC5264625 DOI: 10.1007/s10616-016-0039-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 10/31/2016] [Indexed: 12/21/2022] Open
Abstract
Sodium butyrate (SB) is one of the histone deacetylase inhibitors (HDACi's) that is recently evidenced to have a prooxidant activity and an ability to reduce hydrogen peroxide-induced DNA damage. Since the majority of estrogen receptor positive breast cancer patients are treated with tamoxifen citrate (TC), which exerts well established oxidative and genotoxic effects, thus the basic objective of this study is to determine whether SB could ameliorate or curate tamoxifen citrate-induced oxidative DNA damage and genotoxic effect in vivo through up-regulation of some antioxidant enzymes. The individual and combined effects of SB and TC have been examined on rat bone marrow cells, using Micronucleus assays (MN), Comet assay, DNA fragmentation, expression of some antioxidant genes using Real time-PCR and finally, oxidative stress analysis. SB significantly increased the mitotic activity (P < 0.05), while TC induced marked micronuclei and oxidative DNA damage, in the SB post-treatment group, the combination of SB (300 mg/kg) and TC (40 mg/kg) was able to decrease the induction of MN and oxidative DNA damage through up-regulation of Cat, Sod and Gpx1 genes significantly at (P < 0.05) more efficiently than that in the SB pre-treatment one. Therefore, we postulate that SB can be used therapeutically in combination with TC treatment to modulate TC genotoxic effect by reducing its oxidative stress, and thus being an appropriate agonist agent to combine with TC than each compound alone.
Collapse
|
6
|
Chandra V, Kim JJ, Benbrook DM, Dwivedi A, Rai R. Therapeutic options for management of endometrial hyperplasia. J Gynecol Oncol 2015; 27:e8. [PMID: 26463434 PMCID: PMC4695458 DOI: 10.3802/jgo.2016.27.e8] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 07/24/2015] [Accepted: 07/31/2015] [Indexed: 12/24/2022] Open
Abstract
Endometrial hyperplasia (EH) comprises a spectrum of changes in the endometrium ranging from a slightly disordered pattern that exaggerates the alterations seen in the late proliferative phase of the menstrual cycle to irregular, hyperchromatic lesions that are similar to endometrioid adenocarcinoma. Generally, EH is caused by continuous exposure of estrogen unopposed by progesterone, polycystic ovary syndrome, tamoxifen, or hormone replacement therapy. Since it can progress, or often occur coincidentally with endometrial carcinoma, EH is of clinical importance, and the reversion of hyperplasia to normal endometrium represents the key conservative treatment for prevention of the development of adenocarcinoma. Presently, cyclic progestin or hysterectomy constitutes the major treatment option for EH without or with atypia, respectively. However, clinical trials of hormonal therapies and definitive standard treatments remain to be established for the management of EH. Moreover, therapeutic options for EH patients who wish to preserve fertility are challenging and require nonsurgical management. Therefore, future studies should focus on evaluation of new treatment strategies and novel compounds that could simultaneously target pathways involved in the pathogenesis of estradiol-induced EH. Novel therapeutic agents precisely targeting the inhibition of estrogen receptor, growth factor receptors, and signal transduction pathways are likely to constitute an optimal approach for treatment of EH.
Collapse
Affiliation(s)
- Vishal Chandra
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Jong Joo Kim
- School of Biotechnology, Yeungnam University, Gyeongsan, Korea
| | - Doris Mangiaracina Benbrook
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anila Dwivedi
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rajani Rai
- School of Biotechnology, Yeungnam University, Gyeongsan, Korea.
| |
Collapse
|
7
|
The phenolic acids of Agen prunes (dried plums) or Agen prune juice concentrates do not account for the protective action on bone in a rat model of postmenopausal osteoporosis. Nutr Res 2015; 36:161-73. [PMID: 26574736 DOI: 10.1016/j.nutres.2015.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/12/2015] [Accepted: 10/15/2015] [Indexed: 12/20/2022]
Abstract
Dietary supplementation with dried plum (DP) has been shown to protect against and reverse established osteopenia in ovariectomized rodents. Based on in vitro studies, we hypothesized that DP polyphenols may be responsible for that bone-sparing effect. This study was designed to (1) analyze whether the main phenolic acids of DP control preosteoblast proliferation and activity in vitro; (2) determine if the polyphenolic content of DP or DP juice concentrate is the main component improving bone health in vivo; and (3) analyze whether DP metabolites directly modulate preosteoblast physiology ex vivo. In vitro, we found that neochlorogenic, chlorogenic, and caffeic acids induce the proliferation and repress the alkaline phosphatase activity of primary preosteoblasts in a dose-dependent manner. In vivo, low-chlorogenic acid Agen prunes (AP) enriched with a high-fiber diet and low-chlorogenic acid AP juice concentrate prevented the decrease of total femoral bone mineral density induced by estrogen deficiency in 5-month-old female rats and positively restored the variations of the bone markers osteocalcin and deoxypyridinoline. Ex vivo, we demonstrated that serum from rats fed with low-chlorogenic acid AP enriched with a high-fiber diet showed repressed proliferation and stimulated alkaline phosphatase activity of primary preosteoblasts. Overall, the beneficial action of AP on bone health was not dependent on its polyphenolic content.
Collapse
|
8
|
Thummuri D, Jeengar MK, Shrivastava S, Nemani H, Ramavat RN, Chaudhari P, Naidu VGM. Thymoquinone prevents RANKL-induced osteoclastogenesis activation and osteolysis in an in vivo model of inflammation by suppressing NF-KB and MAPK Signalling. Pharmacol Res 2015; 99:63-73. [PMID: 26022736 DOI: 10.1016/j.phrs.2015.05.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 05/16/2015] [Accepted: 05/17/2015] [Indexed: 01/24/2023]
Abstract
Osteoclasts are multinuclear giant cells responsible for bone resorption in inflammatory bone diseases such as osteoporosis, rheumatoid arthritis and periodontitis. Because of deleterious side effects with currently available drugs the search continues for novel effective and safe therapies. Thymoquinone (TQ), the major bioactive component of Nigella sativa has been investigated for its anti-inflammatory, antioxidant and anticancer activities. However, its effects in osteoclastogenesis have not been reported. In the present study we show for the first time that TQ inhibits nuclear factor-KB ligand (RANKL) induced osteoclastogenesis in RAW 264.7 and primary bone marrow derived macrophages (BMMs) cells. RANKL induced osteoclastogenesis is associated with increased expression of multiple transcription factors via activation of NF-KB, MAPKs signalling and reactive oxygen species (ROS). Mechanistically TQ blocked the RANKL induced NF-KB activation by attenuating the phosphorylation of IkB kinase (IKKα/β). Interestingly, in RAW 264.7 cells TQ inhibited the RANKL induced phosphorylation of MAPKs and mRNA expression of osteoclastic specific genes such as TRAP, DC-STAMP, NFATc1 and c-Fos. In addition, TQ also decreased the RANKL stimulated ROS generation in macropahges (RAW 264.7) and H2O2 induced ROS generation in osteoblasts (MC-3T3-E1). Consistent with in vitro results, TQ inhibited lipopolysaccharide (LPS) induced bone resorption by suppressing the osteoclastogenesis. Indeed, micro-CT analysis showed that bone mineral density (BMD) and bone architecture parameters were positively modulated by TQ. Taken together our data demonstrate that TQ has antiosteoclastogenic effect by inhibiting inflammation induced activation of MAPKs, NF-KB and ROS generation followed by suppressing the gene expression of c-Fos and NFATc1 in osteoclast precursors.
Collapse
Affiliation(s)
- Dinesh Thummuri
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Balanagar, Hyderabad, Telengana 500 037, India
| | - Manish Kumar Jeengar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Balanagar, Hyderabad, Telengana 500 037, India
| | - Shweta Shrivastava
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Balanagar, Hyderabad, Telengana 500 037, India
| | - Harishankar Nemani
- National Centre for Laboratory Animal Sciences, National Institute of Nutrition, Habsiguda, Hyderabad, Telengana 500 037, India
| | - Ravindar Naik Ramavat
- National Centre for Laboratory Animal Sciences, National Institute of Nutrition, Habsiguda, Hyderabad, Telengana 500 037, India
| | - Pradip Chaudhari
- Comparative Oncology Program & Small Animal Imaging Facility, Advanced Center for Treatment, Research & Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410 210, India
| | - V G M Naidu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Balanagar, Hyderabad, Telengana 500 037, India.
| |
Collapse
|
9
|
Thougaard AV, Christiansen J, Mow T, Hornberg JJ. Validation of a high throughput flow cytometric in vitro micronucleus assay including assessment of metabolic activation in TK6 cells. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2014; 55:704-718. [PMID: 25111698 DOI: 10.1002/em.21891] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/22/2014] [Accepted: 07/04/2014] [Indexed: 06/03/2023]
Abstract
Genotoxicity is an unacceptable property for new drug candidates and we employ three screening assays during the drug discovery process to identify genotoxicity early and optimize chemical series. One of these methods is the flow cytometric in vitro micronucleus assay for which protocol optimizations have been described recently. Here, we report further validation of the assay in TK6 cells including assessment of metabolic activation. We first optimized assay conditions to allow for testing with and without metabolic activation in parallel in a 96-well plate format. Then, we tested a set of 48 compounds carefully selected to contain known in vivo genotoxins, nongenotoxins and drugs. Avoidance of irrelevant positives, a known issue with mammalian cell-based genotoxicity assays, is important to prevent early deselection of potentially promising compounds. Therefore, we enriched the validation set with compounds that were previously reported to produce irrelevant positive results in mammalian cell-based genotoxicity assays. The resulting dataset was used to set the relevant cut-off values for scoring a compound positive or negative, such that we obtained an optimal balance of high sensitivity (88%) and high specificity (87%). Finally, we tested an additional set of 16 drugs to further probe assay performance and 14 of them were classified correctly. To our knowledge, the present study is the most comprehensive validation of the in vitro flow cytometric micronucleus assay and the first to report parallel assessment with metabolic activation in reasonable throughput. The assay allows for rapidly screening novel compounds for genotoxicity and is therefore well-suited for use in early drug discovery projects. Environ.
Collapse
Affiliation(s)
- Annemette V Thougaard
- Department of Exploratory Toxicology, H. Lundbeck A/S, Ottiliavej 9, DK-2500, Valby, Denmark
| | | | | | | |
Collapse
|
10
|
Mandelblatt JS, Jacobsen PB, Ahles T. Cognitive effects of cancer systemic therapy: implications for the care of older patients and survivors. J Clin Oncol 2014; 32:2617-26. [PMID: 25071135 PMCID: PMC4129505 DOI: 10.1200/jco.2014.55.1259] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The number of patients with cancer who are age 65 years or older (hereinafter "older") is increasing dramatically. One obvious aspect of cancer care for this group is that they are experiencing age-related changes in multiple organ systems, including the brain, which complicates decisions about systemic therapy and assessments of survivorship outcomes. There is a consistent body of evidence from studies that use neuropsychological testing and neuroimaging that supports the existence of impairment following systemic therapy in selected cognitive domains among some older patients with cancer. Impairment in one or more cognitive domains could have important effects in the daily lives of older patients. However, an imperfect understanding of the precise biologic mechanisms underlying cognitive impairment after systemic treatment precludes development of validated methods for predicting which older patients are at risk. From what is known, risks may include lifestyle factors such as smoking, genetic predisposition, and specific comorbidities such as diabetes and cardiovascular disease. Risk also interacts with physiologic and cognitive reserve, because even at the same chronological age and with the same number of illnesses, older patients vary from having high reserve (ie, biologically younger than their age) to being frail (biologically older than their age). Surveillance for the presence of cognitive impairment is also an important component of long-term survivorship care with older patients. Increasing the workforce of cancer care providers who have geriatrics training or who are working within multidisciplinary teams that have this type of expertise would be one avenue toward integrating assessment of the cognitive effects of cancer systemic therapy into routine clinical practice.
Collapse
Affiliation(s)
- Jeanne S Mandelblatt
- Jeanne S. Mandelblatt, Georgetown University, Washington, DC; Paul B. Jacobsen, Moffitt Cancer Center, Tampa, FL; and Tim Ahles, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY.
| | - Paul B Jacobsen
- Jeanne S. Mandelblatt, Georgetown University, Washington, DC; Paul B. Jacobsen, Moffitt Cancer Center, Tampa, FL; and Tim Ahles, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Tim Ahles
- Jeanne S. Mandelblatt, Georgetown University, Washington, DC; Paul B. Jacobsen, Moffitt Cancer Center, Tampa, FL; and Tim Ahles, Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| |
Collapse
|
11
|
Mustonen MVJ, Pyrhönen S, Kellokumpu-Lehtinen PL. Toremifene in the treatment of breast cancer. World J Clin Oncol 2014; 5:393-405. [PMID: 25114854 PMCID: PMC4127610 DOI: 10.5306/wjco.v5.i3.393] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 04/08/2014] [Accepted: 05/29/2014] [Indexed: 02/06/2023] Open
Abstract
Although more widespread screening and routine adjuvant therapy has improved the outcome for breast cancer patients in recent years, there remains considerable scope for improving the efficacy, safety and tolerability of adjuvant therapy in the early stage disease and the treatment of advanced disease. Toremifene is a selective estrogen receptor modifier (SERM) that has been widely used for decades in hormone receptor positive breast cancer both in early and late stage disease. Its efficacy has been well established in nine prospective randomized phase III trials compared to tamoxifen involving more than 5500 patients, as well as in several large uncontrolled and non-randomized studies. Although most studies show therapeutic equivalence between the two SERMs, some show an advantage for toremifene. Several meta-analyses have also confirmed that the efficacy of toremifene is at least as good as that of tamoxifen. In terms of safety and tolerability toremifene is broadly similar to tamoxifen although there is some evidence that toremifene is less likely to cause uterine neoplasms, serious vascular events and it has a more positive effect on serum lipids than does tamoxifen. Toremifene is therefore effective and safe in the treatment of breast cancer. It provides not only a useful therapeutic alternative to tamoxifen, but may bring specific benefits.
Collapse
|
12
|
Yaacob NS, Ismail NF. Comparison of cytotoxicity and genotoxicity of 4-hydroxytamoxifen in combination with Tualang honey in MCF-7 and MCF-10A cells. Altern Ther Health Med 2014; 14:106. [PMID: 24646375 PMCID: PMC3994783 DOI: 10.1186/1472-6882-14-106] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Accepted: 03/12/2014] [Indexed: 02/07/2023]
Abstract
Background The Malaysian Tualang honey (TH) is not only cytotoxic to human breast cancer cell lines but it has recently been reported to promote the anticancer activity induced by tamoxifen in MCF-7 and MDA-MB-231 cells suggesting its potential as an adjuvant for the chemotherapeutic agent. However, tamoxifen produces adverse effects that could be due to its ability to induce cellular DNA damage. Therefore, the study is undertaken to determine the possible modulation of the activity of 4-hydroxytamoxifen (OHT), an active metabolite of tamoxifen, by TH in non-cancerous epithelial cell line, MCF-10A, in comparison with MCF-7 cells. Methods MCF-7 and MCF-10A cells were treated with TH, OHT or the combination of both and cytotoxicity and antiproliferative activity were determined using LDH and MTT assays, respectively. The effect on cellular DNA integrity was analysed by comet assay and the expression of DNA repair enzymes was determined by Western blotting. Results OHT exposure was cytotoxic to both cell lines whereas TH was cytotoxic to MCF-7 cells only. TH also significantly decreased the cytotoxic effect of OHT in MCF-10A but not in MCF-7 cells. TH induced proliferation of MCF10A cells but OHT caused growth inhibition that was abrogated by the concomitant treatment with TH. While TH enhanced the OHT-induced DNA damage in the cancer cells, it dampened the genotoxic effect of OHT in the non-cancerous cells. This was supported by the increased expression of DNA repair proteins, Ku70 and Ku80, in MCF-10A cells by TH. Conclusion The findings indicate that TH could afford protection of non-cancerous cells from the toxic effects of tamoxifen by increasing the efficiency of DNA repair mechanism in these cells.
Collapse
|
13
|
Hernandez-Ramon EE, Sandoval NA, John K, Cline JM, Wood CE, Woodward RA, Poirier MC. Tamoxifen-DNA adduct formation in monkey and human reproductive organs. Carcinogenesis 2014; 35:1172-6. [PMID: 24501327 DOI: 10.1093/carcin/bgu029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The estrogen analog tamoxifen (TAM), used for adjuvant therapy of breast cancer, induces endometrial and uterine tumors in breast cancer patients. Proliferation stimulus of the uterine endometrium is likely involved in tumor induction, but genotoxicity may also play a role. Formation of TAM-DNA adducts in human tissues has been reported but remains controversial. To address this issue, we examined TAM-DNA adducts in uteri from two species of monkeys, Erythrocebus patas (patas) and Macaca fascicularis (macaque), and in human endometrium and myometrium. Monkeys were given 3-4 months of chronic TAM dosing scaled to be equivalent to the daily human dose. In the uteri, livers and brains from the patas (n = 3), and endometrium from the macaques (n = 4), TAM-DNA adducts were measurable by TAM-DNA chemiluminescence immunoassay. Average TAM-DNA adduct values for the patas uteri (23 adducts/10(8) nucleotides) were similar to those found in endometrium of the macaques (19 adducts/10(8) nucleotides). Endometrium of macaques exposed to both TAM and low-dose estradiol (n = 5) averaged 34 adducts/10(8) nucleotides. To examine TAM-DNA persistence in the patas, females (n = 3) were exposed to TAM for 3 months and to no drug for an additional month, resulting in low or non-detectable TAM-DNA in livers and uteri. Human endometrial and myometrial samples from women receiving (n = 8) and not receiving (n = 8) TAM therapy were also evaluated. Women receiving TAM therapy averaged 10.3 TAM-DNA adducts/10(8) nucleotides, whereas unexposed women showed no detectable TAM-DNA. The data indicate that genotoxicity, in addition to estrogen agonist effects, may contribute to TAM-induced human endometrial cancer.
Collapse
Affiliation(s)
- Elena E Hernandez-Ramon
- Carcinogen-DNA Interactions Section, LCBG, CCR, National Cancer Institute, NIH, Building 37, Room 4032, NIH 37 Convent Drive, MSC-4255, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Pottenger LH, Andrews LS, Bachman AN, Boogaard PJ, Cadet J, Embry MR, Farmer PB, Himmelstein MW, Jarabek AM, Martin EA, Mauthe RJ, Persaud R, Preston RJ, Schoeny R, Skare J, Swenberg JA, Williams GM, Zeiger E, Zhang F, Kim JH. An organizational approach for the assessment of DNA adduct data in risk assessment: case studies for aflatoxin B1, tamoxifen and vinyl chloride. Crit Rev Toxicol 2014; 44:348-91. [DOI: 10.3109/10408444.2013.873768] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
15
|
Evaluating the potential bioactivity of a novel compound ER1626. PLoS One 2014; 9:e86509. [PMID: 24475135 PMCID: PMC3903524 DOI: 10.1371/journal.pone.0086509] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 12/10/2013] [Indexed: 12/21/2022] Open
Abstract
Background ER1626, a novel compound, is a derivate of indeno-isoquinoline ketone. This study was designed to evaluate the biological activity and potential anti-tumor mechanism of ER1626. Method MTT assay, scratch assay and flow cytometry were used to determine cell proliferation, cell migration and cell cycle distribution as well as cell apoptosis on human breast cancer MCF-7 cells and endometrial cancer Ishikawa cells. We also explored the antiangiogenic effect of ER1626 on HUVEC cells and chicken embryos. The expression of estrogen receptor protein was investigated with western-blot analysis. Results ER1626 down-regulated the expression of estrogen receptor α protein and up-regulated β protein in MCF-7 and Ishikawa cells. The value of IC50 of ER1626 on MCF-7 and Ishikawa cells were respectively 8.52 and 3.08 µmol/L. Meanwhile, ER1626 decreased VEGF secretion of MCF-7 and Ishikawa cells, disturbed the formation of VEGF-stimulated tubular structure in HUVEC cells, and inhibited the angiogenesis on the chicken chorioallantoic membrane. Scratch assay revealed that ER1626 suppressed the migration of MCF-7, Ishikawa and HUVEC cells. In addition to induction tumor cell apoptosis, ER1626 arrested cell cycle in G1/G0 phase in MCF-7 cells and G2/M phase in Ishikawa cells. Conclusion In conclusion, our results demonstrated that ER1626 has favorable bioactivities to be a potential candidate against breast cancer and angiogenesis.
Collapse
|
16
|
Mandelblatt JS, Hurria A, McDonald BC, Saykin AJ, Stern RA, VanMeter JW, McGuckin M, Traina T, Denduluri N, Turner S, Howard D, Jacobsen PB, Ahles T. Cognitive effects of cancer and its treatments at the intersection of aging: what do we know; what do we need to know? Semin Oncol 2013; 40:709-25. [PMID: 24331192 PMCID: PMC3880205 DOI: 10.1053/j.seminoncol.2013.09.006] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
There is a fairly consistent, albeit non-universal body of research documenting cognitive declines after cancer and its treatments. While few of these studies have included subjects aged 65 years and older, it is logical to expect that older patients are at risk of cognitive decline. Here, we use breast cancer as an exemplar disease for inquiry into the intersection of aging and cognitive effects of cancer and its therapies. There are a striking number of common underlying potential biological risks and pathways for the development of cancer, cancer-related cognitive declines, and aging processes, including the development of a frail phenotype. Candidate shared pathways include changes in hormonal milieu, inflammation, oxidative stress, DNA damage and compromised DNA repair, genetic susceptibility, decreased brain blood flow or disruption of the blood-brain barrier, direct neurotoxicity, decreased telomere length, and cell senescence. There also are similar structure and functional changes seen in brain imaging studies of cancer patients and those seen with "normal" aging and Alzheimer's disease. Disentangling the role of these overlapping processes is difficult since they require aged animal models and large samples of older human subjects. From what we do know, frailty and its low cognitive reserve seem to be a clinically useful marker of risk for cognitive decline after cancer and its treatments. This and other results from this review suggest the value of geriatric assessments to identify older patients at the highest risk of cognitive decline. Further research is needed to understand the interactions between aging, genetic predisposition, lifestyle factors, and frailty phenotypes to best identify the subgroups of older patients at greatest risk for decline and to develop behavioral and pharmacological interventions targeting this group. We recommend that basic science and population trials be developed specifically for older hosts with intermediate endpoints of relevance to this group, including cognitive function and trajectories of frailty. Clinicians and their older patients can advance the field by active encouragement of and participation in research designed to improve the care and outcomes of the growing population of older cancer patients.
Collapse
Affiliation(s)
- Jeanne S Mandelblatt
- Departments of Oncology and Population Sciences, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC.
| | - Arti Hurria
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Brenna C McDonald
- Center for Neuroimaging, Department of Radiology and Imaging Sciences and the Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN
| | - Andrew J Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences and the Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN
| | - Robert A Stern
- Departments of Neurology and Neurosurgery and Director, Clinical Core, BU Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA
| | - John W VanMeter
- Department of Neurology, Georgetown University Medical Center, Georgetown University, Washington, DC
| | - Meghan McGuckin
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Tiffani Traina
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Neelima Denduluri
- Department of Medicine, Georgetown University; Virginia Cancer Specialists, US Oncology, Arlington, VA
| | - Scott Turner
- Department of Neurology, Georgetown University Medical Center, Georgetown University, Washington, DC
| | - Darlene Howard
- Department of Psychology, Georgetown University, Washington, DC
| | - Paul B Jacobsen
- Division of Population Science, Moffitt Cancer Center, Tampa, FL
| | - Tim Ahles
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan-Kettering Cancer Center, New York, NY; Department of Psychiatry, Weill Cornell Medical College, New York, NY
| |
Collapse
|
17
|
Léotoing L, Wauquier F, Guicheux J, Miot-Noirault E, Wittrant Y, Coxam V. The polyphenol fisetin protects bone by repressing NF-κB and MKP-1-dependent signaling pathways in osteoclasts. PLoS One 2013; 8:e68388. [PMID: 23861901 PMCID: PMC3701685 DOI: 10.1371/journal.pone.0068388] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 06/03/2013] [Indexed: 11/18/2022] Open
Abstract
Osteoporosis is a bone pathology leading to increase fractures risk and challenging quality of life. Since current treatments could exhibit deleterious side effects, the use of food compounds derived from plants represents a promising innovative alternative due to their potential therapeutic and preventive activities against human diseases. In this study, we investigated the ability of the polyphenol fisetin to counter osteoporosis and analyzed the cellular and molecular mechanisms involved. In vivo, fisetin consumption significantly prevented bone loss in estrogen deficiency and inflammation mice osteoporosis models. Indeed, bone mineral density, micro-architecture parameters and bone markers were positively modulated by fisetin. Consistent with in vivo results, we showed that fisetin represses RANKL-induced osteoclast differentiation and activity as demonstrated by an inhibition of multinucleated cells formation, TRAP activity and differentiation genes expression. The signaling pathways NF-κB, p38 MAPK, JNK and the key transcription factors c-Fos and NFATc1 expressions induced by RANKL, were negatively regulated by fisetin. We further showed that fisetin inhibits the constitutive proteasomal degradation of MKP-1, the phosphatase that deactivates p38 and JNK. Consistently, using shRNA stable cell lines, we demonstrated that impairment of MKP-1 decreases fisetin potency. Taken together, these results strongly support that fisetin should be further considered as a bone protective agent.
Collapse
Affiliation(s)
- Laurent Léotoing
- Clermont Université, Université d'Auvergne, Clermont-Ferrand, France
- INRA, UMR 1019, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France
| | - Fabien Wauquier
- Clermont Université, Université d'Auvergne, Clermont-Ferrand, France
- INRA, UMR 1019, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France
| | - Jérôme Guicheux
- Université de Nantes, UFR Odontologie, Nantes, France
- NSERM, UMRS 791, LIOAD, Nantes, France
| | - Elisabeth Miot-Noirault
- Clermont Université, Université d'Auvergne, Clermont-Ferrand, France
- INSERM, UMR 990, Clermont-Ferrand, France
| | - Yohann Wittrant
- Clermont Université, Université d'Auvergne, Clermont-Ferrand, France
- INRA, UMR 1019, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France
| | - Véronique Coxam
- Clermont Université, Université d'Auvergne, Clermont-Ferrand, France
- INRA, UMR 1019, Unité de Nutrition Humaine, CRNH Auvergne, Clermont-Ferrand, France
- * E-mail:
| |
Collapse
|
18
|
Dias MC, Furtado KS, Rodrigues MAM, Barbisan LF. Effects of Ginkgo biloba on chemically-induced mammary tumors in rats receiving tamoxifen. Altern Ther Health Med 2013; 13:93. [PMID: 23634930 PMCID: PMC3655872 DOI: 10.1186/1472-6882-13-93] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 04/24/2013] [Indexed: 11/26/2022]
Abstract
Background Ginkgo biloba extract (GbE) is used extensively by breast cancer patients undergoing treatment with Tamoxifen (TAM). Thus, the present study investigated the effects of GbE in female Sprague–Dawley (SD) rats bearing chemically-induced mammary tumors and receiving TAM. Methods Animals bearing mammary tumors (≥1 cm in diameter) were divided into four groups: TAM [10 mg/kg, intragastrically (i.g.)], TAM plus GbE [50 and 100 mg/kg, intraperitoneally (i.p.)] or an untreated control group. After 4 weeks, the therapeutic efficacy of the different treatments was evaluated by measuring the tumor volume (cm3) and the proportions of each tumor that were alive, necrotic or degenerative (mm2). In addition, labeling indexes (LI%) were calculated for cell proliferation (PCNA LI%) and apoptosis (cleaved caspase-3 LI%), expression of estrogen receptor-alpha (ER-α) and p63 biomarkers. Results Overall, the tumor volume and the PCNA LI% within live tumor areas were reduced by 83% and 99%, respectively, in all TAM-treated groups when compared to the untreated control group. GbE treatment (100 mg/kg) reduced the proportions of live (24.8%) and necrotic areas (2.9%) (p = 0.046 and p = 0.038, respectively) and significantly increased the proportion of degenerative areas (72.9%) (p = 0.004) in mammary tumors when compared to the group treated only with TAM. The expression of ER-α, p63 and cleaved caspase-3 in live tumor tissues was not modified by GbE treatment. Conclusions Co-treatment with 100 mg/kg GbE presented a slightly beneficial effect on the therapeutic efficacy of TAM in female SD rats bearing mammary tumors.
Collapse
|
19
|
Ahles TA. Brain vulnerability to chemotherapy toxicities. Psychooncology 2012; 21:1141-8. [PMID: 23023994 DOI: 10.1002/pon.3196] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 09/06/2012] [Indexed: 12/21/2022]
Abstract
Chemotherapy-induced cognitive changes have been an increasing concern among cancer survivors. By using adjuvant treatment for breast cancer as the prototype, this manuscript reviews research from neuropsychological, imaging, genetic, and animal model studies that have examined the clinical presentation and potential mechanisms for cognitive changes associated with exposure to chemotherapy. An impressive body of research supports the hypothesis that a subgroup of patients is vulnerable to post-treatment cognitive changes, although not exclusively related to chemotherapy. Further, imaging and animal model studies provide accumulating evidence of putative mechanisms for chemotherapy-induced cognitive change. Models of aging are also reviewed in support of the proposal that cognitive changes associated with cancer and cancer treatments can be viewed in the context of factors that affect the trajectory of normal aging.
Collapse
Affiliation(s)
- Tim A Ahles
- Neurocognitive Research Laboratory, Department of Psychiatry and Behavioral Sciences, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
20
|
Ahles TA, Root JC, Ryan EL. Cancer- and cancer treatment-associated cognitive change: an update on the state of the science. J Clin Oncol 2012; 30:3675-86. [PMID: 23008308 DOI: 10.1200/jco.2012.43.0116] [Citation(s) in RCA: 492] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Cognitive changes associated with cancer and cancer treatments have become an increasing concern. Using breast cancer as the prototype, we reviewed the research from neuropsychological, imaging, genetic, and animal studies that have examined pre- and post-treatment cognitive change. An impressive body of research supports the contention that a subgroup of patients is vulnerable to post-treatment cognitive problems. We also propose that models of aging may be a useful conceptual framework for guiding research in this area and suggest that a useful perspective may be viewing cognitive change in patients with cancer within the context of factors that influence the trajectory of normal aging.
Collapse
Affiliation(s)
- Tim A Ahles
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan-Kettering Cancer Center, New York, NY 10022, USA.
| | | | | |
Collapse
|
21
|
Lee YM, Lee JY, Ho CC, Hong QS, Yu SL, Tzeng CR, Yang PC, Chen HW. miRNA-34b as a tumor suppressor in estrogen-dependent growth of breast cancer cells. Breast Cancer Res 2011; 13:R116. [PMID: 22113133 PMCID: PMC3326558 DOI: 10.1186/bcr3059] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 07/06/2011] [Accepted: 11/23/2011] [Indexed: 02/06/2023] Open
Abstract
Introduction Estrogen is involved in several physiological and pathological processes through estrogen receptor (ER)-mediated transcriptional gene regulation. miRNAs (miRs), which are noncoding RNA genes, may respond to estrogen and serve as posttranscriptional regulators in tumorigenic progression, especially in breast cancer; however, only limited information about this possibility is available. In the present study, we identified the estrogen-regulated miR-34b and investigated its functional role in breast cancer progression. Methods Estrogen-regulated miRNAs were identified by using a TaqMan low density array. Our in vivo Tet-On system orthotopic model revealed the tumor-suppressive ability of miR-34b. Luciferase reporter assays and chromatin immunoprecipitation assay demonstrated miR-34b were regulated by p53-ER interaction. Results In this study, we identified one such estrogen downregulated miRNA, miR-34b, as an oncosuppressor that targets cyclin D1 and Jagged-1 (JAG1) in an ER+/wild-type p53 breast cancer cell line (MCF-7), as well as in ovarian and endometrial cells, but not in ER-negative or mutant p53 breast cancer cell lines (T47D, MBA-MB-361 and MDA-MB-435). There is a negative association between ERα and miR-34b expression levels in ER+ breast cancer patients. Tet-On induction of miR-34b can cause inhibition of tumor growth and cell proliferation. Also, the overexpression of miR-34b inhibited ER+ breast tumor growth in an orthotopic mammary fat pad xenograft mouse model. Further validation indicated that estrogen's inhibition of miR-34b expression was mediated by interactions between ERα and p53, not by DNA methylation regulation. The xenoestrogens diethylstilbestrol and zeranol also showed similar estrogenic effects by inhibiting miR-34b expression and by restoring the protein levels of the miR-34b targets cyclin D1 and JAG1 in MCF-7 cells. Conclusions These findings reveal that miR-34b is an oncosuppressor miRNA requiring both ER+ and wild-type p53 phenotypes in breast cancer cells. These results improve our ability to develop new therapeutic strategies to target the complex estrogenic pathway in human breast cancer progression through miRNA regulation.
Collapse
Affiliation(s)
- Yee-Ming Lee
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, No, 1, Sec, 1, Ren-Ai Road, Taipei, 100, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Hepatocellular carcinoma arising from hepatocellular adenoma in a hepatitis B virus-associated cirrhotic liver. Clin Radiol 2011; 67:329-33. [PMID: 22079485 DOI: 10.1016/j.crad.2011.09.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 08/31/2011] [Accepted: 09/14/2011] [Indexed: 02/06/2023]
Abstract
Hepatocellular adenoma (HCA) is a rare, benign proliferation of hepatocytes that occurs mostly in a normal liver and in extreme rare cases, occurs in a cirrhotic liver. Hepatocellular carcinomas (HCC) arising within HCA through malignant transformation is rare. The specific incidence and mechanism of malignant transformation has not been established, but the long term use of oral contraceptives is considered a causative agent. We report a case of HCC arising from HCA detected in a hepatitis B-related cirrhotic liver with serial radiologic images.
Collapse
|
23
|
Williams-Brown MY, Salih SM, Xu X, Veenstra TD, Saeed M, Theiler SK, Diaz-Arrastia CR, Salama SA. The effect of tamoxifen and raloxifene on estrogen metabolism and endometrial cancer risk. J Steroid Biochem Mol Biol 2011; 126:78-86. [PMID: 21600284 PMCID: PMC3421458 DOI: 10.1016/j.jsbmb.2011.05.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 03/22/2011] [Accepted: 05/03/2011] [Indexed: 11/18/2022]
Abstract
Selective estrogen receptor modulators (SERMs) demonstrate differential endometrial cancer (EC) risk. While tamoxifen (TAM) use increases the risk of endometrial hyperplasia and malignancy, raloxifene (RAL) has neutral effects on the uterus. How TAM increases the risk of EC and why TAM and RAL differentially modulate the risk for EC, however, remain elusive. Here, we tested the hypothesis that TAM increases the risk for EC, at least in part, by enhancing the local estrogen biosynthesis and directing estrogen metabolism towards the formation of genotoxic and hormonally active estrogen metabolites. In addition, the differential effects of TAM and RAL in EC risk are attributed to their differential effect on estrogen metabolism/metabolites. The endometrial cancer cell line (Ishikawa cells) and the nonmalignant immortalized human endometrial glandular cell line (EM1) were used for the study. The profile of estrogen/estrogen metabolites (EM), depurinating estrogen-DNA adducts, and the expression of estrogen-metabolizing enzymes in cells treated with 17β-estradiol (E2) alone or in combination with TAM or RAL were investigated using high performance liquid chromatography-electrospray ionization-tandem mass spectrometry (HPLC-ESI-MS(2)), ultraperformance liquid chromatography/tandem mass spectrometry (UPLC-MS/MS), and Western blot analysis, respectively. TAM significantly increased the total EM and enhanced the formation of hormonally active and carcinogenic estrogen metabolites, 4-hydroxestrone (4-OHE1) and 16α-hydroxyestrone, with concomitant reduction in the formation of antiestrogenic and anticarcinogenic 2-hydroxyestradiol and 2-methoxyestradiol. Furthermore, TAM increased the formation of depurinating estrogen-DNA adducts 4-OHE1 [2]-1-N7Guanine and 4-OHE1 [2]-1-N3 Adenine. TAM-induced alteration in EM and depurinating DNA adduct formation is associated with altered expression of estrogen metabolizing enzymes CYP1A1, CYP1B1, COMT, NQO1, and SF-1 as revealed by Western blot analysis. In contrast to TAM, RAL has minimal effect on EM, estrogen-DNA adduct formation, or estrogen-metabolizing enzymes expression. These data show that TAM perturbs the balance of estrogen-metabolizing enzymes and alters the disposition of estrogen metabolites, which can explain, at least in part, the mechanism for TAM-induced EC. These results also implicate the differential effect of TAM and RAL on estrogen metabolism/metabolites as a potential mechanism for their disparate effects on the endometrium.
Collapse
Affiliation(s)
- Marian Y Williams-Brown
- Department of Obstetrics & Gynecology, University of Texas Medical Branch, Galveston, United States.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Suzuki N, Liu X, Laxmi YRS, Okamoto K, Kim HJ, Zhang G, Chen JJ, Okamoto Y, Shibutani S. Anti-breast cancer potential of SS5020, a novel benzopyran antiestrogen. Int J Cancer 2011; 128:974-82. [PMID: 20824696 PMCID: PMC3011858 DOI: 10.1002/ijc.25659] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2010] [Accepted: 08/26/2010] [Indexed: 12/21/2022]
Abstract
Treatment with tamoxifen (TAM) increases the risk of developing endometrial cancer in women. The carcinogenic effect is thought to involve initiation and/or promotion resulting from DNA damage induced by TAM as well as its estrogenic action. To minimize this serious side-effect while increasing the anti-breast cancer potential, a new benzopyran antiestrogen, 2E-3-{4-[(7-hydroxy-2-oxo-3-phenyl-2H-chromen-4-yl)-methyl]-phenyl}-acrylic acid (SS5020), was synthesized. Unlike TAM, SS5020 exhibits no genotoxic activity to damage DNA. Furthermore, SS5020 does not present significant uterotrophic potential in rats; in contrast, the structurally related compounds, TAM, toremifene, raloxifene (RAL) and SP500263 all have uterotrophic activity. At the human equivalent molar dose of TAM (0.33 or 1.0 mg/kg), SS5020 had much stronger antitumor potential than those same antiestrogens against 7,12-dimethylbenz(a)anthracene-induced mammary carcinoma in rats. The growth of human MCF-7 breast cancer xenograft implanted into athymic nude mice was also effectively suppressed by SS5020. SS5020, lacking genotoxic and estrogenic actions, could be a safer and stronger antiestrogen alternative to TAM and RAL for breast cancer therapy and prevention.
Collapse
Affiliation(s)
- Naomi Suzuki
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - Xiaoping Liu
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - Y. R. Santosh Laxmi
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - Kanako Okamoto
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - Hyo Jeong Kim
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - Guangxiang Zhang
- Department of Preventive Medicine, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - John J. Chen
- Department of Preventive Medicine, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - Yoshinori Okamoto
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| | - Shinya Shibutani
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, New York 11794-8651
| |
Collapse
|
25
|
Hashizume T, Yoshitomi S, Asahi S, Uematsu R, Matsumura S, Chatani F, Oda H. Advantages of Human Hepatocyte-Derived Transformants Expressing a Series of Human Cytochrome P450 Isoforms for Genotoxicity Examination. Toxicol Sci 2010; 116:488-97. [DOI: 10.1093/toxsci/kfq154] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|