Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 161
Filter
1.
Anticancer Drugs ; 35(4): 371-376, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38241197

ABSTRACT

To investigate the effects of tamoxifen (TAM) and toremifene (TOR) on hepatic function and serum lipid levels in breast cancer patients receiving adjuvant endocrine therapy. The clinical data of 597 early breast cancer patients treated at the First Affiliated Hospital of Nanjing Medical University between January 2016 and December 2022 were collected. All the patients received standard adjuvant endocrine therapy with TAM or TOR after chemotherapy. Hepatic function and serum lipid data of all patients before and at 6 months and 1, 2, and 3 years after the treatment were collected retrospectively and analyzed statistically. There: no negative effect on hepatic function was observed in patients treated with either TAM or TOR. The triglyceride levels in both groups increased during treatment, and the effect of TAM on improving total cholesterol levels was stronger. Total cholesterol levels were not affected by time or treatment regimen. The low-density lipoprotein cholesterol levels decreased in both groups, and the effect was similar between groups. TAM can decrease the high-density lipoprotein cholesterol levels, whereas TOR can increase the high-density lipoprotein cholesterol levels, and there was a significant difference between groups. In the postoperative adjuvant endocrine therapy, TOR and TAM will not negatively impact the hepatic function of breast cancer patients, and TOR is better than TAM in the management of serum lipids; therefore, it may be a better choice for clinical medication.


Subject(s)
Breast Neoplasms , Toremifene , Humans , Female , Toremifene/therapeutic use , Toremifene/pharmacology , Tamoxifen/pharmacology , Retrospective Studies , Antineoplastic Agents, Hormonal/adverse effects , Chemotherapy, Adjuvant , Adjuvants, Immunologic , Lipids/therapeutic use , Cholesterol , Lipoproteins, HDL/therapeutic use
2.
J Cancer Res Clin Oncol ; 149(10): 7837-7848, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37036506

ABSTRACT

PURPOSE: Toremifene (TOR) is widely used as an antineoplastic drug and has an inhibitory effect on angiogenesis in mesenteric desmoid tumors and vascular intracranial solitary fibrous tumors. However, no study has investigated the direct effect of TOR on vascular cells. This study aimed at exploring the effect of TOR on the behaviors of vascular smooth muscle cells (VSMCs). METHODS: Human aortic umbilical vascular smooth muscle cells (HAVSMCs) were treated by TOR. Cell morphology, migration, adhesion, and proliferation assay were investigated. The cell cycle, apoptosis, mitochondrial membrane potential, and reactive oxygen species were assessed using flow cytometry. Caspase-3 and 9 activities were assayed using Caspase-3 and Caspase-9 Activity Assay kits, respectively. Immunofluorescence and Western blot assays were carried out to characterize protein expressions of PCNA, p53, and Rho/ROCK signaling pathway. RESULTS: TOR damaged cytoskeleton, inhibited VSMC proliferation, migration, and adhesion, and induced abnormal cell morphology and apoptosis. The antiproliferative activity of TOR was associated with the induction of G0/G1 phase arrest, blocking the cell cycle. TOR disrupted intracellular reactive oxygen species and mitochondrial membrane potential, and enhanced p53 expression and the activities of caspase-3 and caspase-9. Thus, TOR-induced apoptosis by the mitochondrial signaling pathway. Additionally, TOR induced decreased Rho, ROCK, MLC, and pMLC proteins. Collectively, TOR may affect multiple behaviors of VSMCs by damaging cytoskeleton through the Rho/ROCK pathway. CONCLUSION: The adverse effect of TOR on VSMCs could be considered as an important aspect of tumor growth inhibition.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Cell Proliferation , Muscle, Smooth, Vascular/metabolism , Toremifene/metabolism , Toremifene/pharmacology , Caspase 3/metabolism , Caspase 9/metabolism , Caspase 9/pharmacology , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Movement , Antineoplastic Agents/adverse effects , Neoplasms/metabolism , Cells, Cultured
3.
Endocrinology ; 162(10)2021 10 01.
Article in English | MEDLINE | ID: mdl-34265069

ABSTRACT

Cancer-induced skeletal muscle defects show sex-specific differences in severity with men performing poorly compared to women. Hormones and sex chromosomal differences are suggested to mediate these differences, but the functional skeletal muscle markers to document these differences are unknown. We show that the myogenic microRNA miR-486 is a marker of sex-specific differences in cancer-induced skeletal muscle defects. Cancer-induced loss of circulating miR-486 was more severe in men with bladder, lung, and pancreatic cancers compared to women with the same cancer types. In a syngeneic model of pancreatic cancer, circulating and skeletal muscle loss of miR-486 was more severe in male mice compared to female mice. Estradiol (E2) and the clinically used selective estrogen receptor modulator toremifene increased miR-486 in undifferentiated and differentiated myoblast cell line C2C12 and E2-inducible expression correlated with direct binding of estrogen receptor alpha (ERα) to the regulatory region of the miR-486 gene. E2 and toremifene reduced the actions of cytokines such as myostatin, transforming growth factor ß, and tumor necrosis factor α, which mediate cancer-induced skeletal muscle wasting. E2- and toremifene-treated C2C12 myoblast/myotube cells contained elevated levels of active protein kinase B (AKT) with a corresponding decrease in the levels of its negative regulator PTEN, which is a target of miR-486. We propose an ERα:E2-miR-486-AKT signaling axis, which reduces the deleterious effects of cancer-induced cytokines/chemokines on skeletal muscle mass and/or function.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/biosynthesis , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Neoplasms/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Estradiol/pharmacology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Muscular Diseases/complications , Myostatin/biosynthesis , Neoplasms/complications , Sex Factors , Signal Transduction , Toremifene/pharmacology , Transforming Growth Factor beta/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
4.
BMC Cancer ; 21(1): 798, 2021 Jul 10.
Article in English | MEDLINE | ID: mdl-34246237

ABSTRACT

BACKGROUND: Tamoxifen (TAM) and Toremifene (TOR), two kinds of selective estrogen receptor modulators (SERMs), have equal efficacy in breast cancer patients. However, TAM has been proved to affect serum lipid profiles and cause fatty liver disease. The study aimed to compare the effects of TAM and TOR on fatty liver development and lipid profiles. METHODS: This study performed a retrospective analysis of 308 SERMs-treated early breast cancer patients who were matched 1:1 based on propensity scores. The follow-up period was 3 years. The primary outcomes were fatty liver detected by ultrasonography or computed tomography (CT), variation in fibrosis indexes, and serum lipid profiles change. RESULTS: The cumulative incidence rate of new-onset fatty liver was higher in the TAM group than in the TOR group (113.2 vs. 67.2 per 1000 person-years, p < 0.001), and more severe fatty livers occurred in the TAM group (25.5 vs. 7.5 per 1000 person-years, p = 0.003). According to the Kaplan-Meier curves, TAM significantly increased the risk of new-onset fatty liver (25.97% vs. 17.53%, p = 0.0243) and the severe fatty liver (5.84% vs. 1.95%, p = 0.0429). TOR decreased the risk of new-onset fatty liver by 45% (hazard ratio = 0.55, p = 0.020) and showed lower fibrotic burden, independent of obesity, lipid, and liver enzyme levels. TOR increased triglycerides less than TAM, and TOR increased high-density lipoprotein cholesterol, while TAM did the opposite. No significant differences in total cholesterol and low-density lipoprotein cholesterol are observed between the two groups. CONCLUSIONS: TAM treatment is significantly associated with more severe fatty liver disease and liver fibrosis, while TOR is associated with an overall improvement in lipid profiles, which supports continuous monitoring of liver imaging and serum lipid levels during SERM treatment.


Subject(s)
Breast Neoplasms/drug therapy , Fatty Liver/drug therapy , Lipids/blood , Tamoxifen/therapeutic use , Toremifene/therapeutic use , Adult , Breast Neoplasms/pathology , Female , Humans , Middle Aged , Retrospective Studies , Tamoxifen/pharmacology , Toremifene/pharmacology
5.
J Virol ; 94(18)2020 08 31.
Article in English | MEDLINE | ID: mdl-32611759

ABSTRACT

Ebola virus (EBOV) entry into cells is mediated by its spike glycoprotein (GP). Following attachment and internalization, virions traffic to late endosomes where GP is cleaved by host cysteine proteases. Cleaved GP then binds its cellular receptor, Niemann-Pick C1. In response to an unknown cellular trigger, GP undergoes conformational rearrangements that drive fusion of viral and endosomal membranes. The temperature-dependent stability (thermostability) of the prefusion conformers of class I viral fusion glycoproteins, including those of filovirus GPs, has provided insights into their propensity to undergo fusion-related rearrangements. However, previously described assays have relied on soluble glycoprotein ectodomains. Here, we developed a simple enzyme-linked immunosorbent assay (ELISA)-based assay that uses the temperature-dependent loss of conformational epitopes to measure thermostability of GP embedded in viral membranes. The base and glycan cap subdomains of all filovirus GPs tested suffered a concerted loss of prefusion conformation at elevated temperatures but did so at different temperature ranges, indicating virus-specific differences in thermostability. Despite these differences, all of these GPs displayed reduced thermostability upon cleavage to GP conformers (GPCL). Surprisingly, acid pH enhanced, rather than decreased, GP thermostability, suggesting it could enhance viral survival in hostile endo/lysosomal compartments. Finally, we confirmed and extended previous findings that some small-molecule inhibitors of filovirus entry destabilize EBOV GP and uncovered evidence that the most potent inhibitors act through multiple mechanisms. We establish the epitope-loss ELISA as a useful tool for studies of filovirus entry, engineering of GP variants with enhanced stability for use in vaccine development, and discovery of new stability-modulating antivirals.IMPORTANCE The development of Ebola virus countermeasures is challenged by our limited understanding of cell entry, especially at the step of membrane fusion. The surface-exposed viral protein, GP, mediates membrane fusion and undergoes major structural rearrangements during this process. The stability of GP at elevated temperatures (thermostability) can provide insights into its capacity to undergo these rearrangements. Here, we describe a new assay that uses GP-specific antibodies to measure GP thermostability under a variety of conditions relevant to viral entry. We show that proteolytic cleavage and acid pH have significant effects on GP thermostability that shed light on their respective roles in viral entry. We also show that the assay can be used to study how small-molecule entry inhibitors affect GP stability. This work provides a simple and readily accessible assay to engineer stabilized GP variants for antiviral vaccines and to discover and improve drugs that act by modulating GP stability.


Subject(s)
Ebolavirus/drug effects , Niemann-Pick C1 Protein/antagonists & inhibitors , Receptors, Virus/antagonists & inhibitors , Viral Envelope Proteins/antagonists & inhibitors , Viral Fusion Proteins/antagonists & inhibitors , Virion/drug effects , Animals , Binding Sites , Biological Assay , Chlorocebus aethiops , Clomiphene/chemistry , Clomiphene/pharmacology , Ebolavirus/chemistry , Ebolavirus/genetics , Ebolavirus/metabolism , Epitopes/chemistry , Epitopes/genetics , Epitopes/metabolism , Hot Temperature , Hydrogen-Ion Concentration , Molecular Docking Simulation , Niemann-Pick C1 Protein/chemistry , Niemann-Pick C1 Protein/genetics , Niemann-Pick C1 Protein/metabolism , Protein Binding/drug effects , Protein Interaction Domains and Motifs , Protein Stability , Protein Structure, Tertiary , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/chemistry , Tamoxifen/pharmacology , Toremifene/chemistry , Toremifene/pharmacology , Vero Cells , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Virion/chemistry , Virion/genetics , Virion/metabolism
6.
J Med Chem ; 63(13): 7211-7225, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32490678

ABSTRACT

The recent Ebola epidemics in West Africa underscore the great need for effective and practical therapies for future Ebola virus outbreaks. We have discovered a new series of remarkably potent small molecule inhibitors of Ebola virus entry. These 4-(aminomethyl)benzamide-based inhibitors are also effective against Marburg virus. Synthetic routes to these compounds allowed for the preparation of a wide variety of structures, including a conformationally restrained subset of indolines (compounds 41-50). Compounds 20, 23, 32, 33, and 35 are superior inhibitors of Ebola (Mayinga) and Marburg (Angola) infectious viruses. Representative compounds (20, 32, and 35) have shown good metabolic stability in plasma and liver microsomes (rat and human), and 32 did not inhibit CYP3A4 nor CYP2C9. These 4-(aminomethyl)benzamides are suitable for further optimization as inhibitors of filovirus entry, with the potential to be developed as therapeutic agents for the treatment and control of Ebola virus infections.


Subject(s)
Antiviral Agents/pharmacology , Benzamides/pharmacology , Hemorrhagic Fever, Ebola/virology , Marburg Virus Disease/virology , Virus Internalization/drug effects , A549 Cells , Animals , Antiviral Agents/chemistry , Benzamides/chemistry , Chlorocebus aethiops , Cytochrome P-450 CYP3A Inhibitors/chemistry , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Evaluation, Preclinical , Humans , Microsomes, Liver/drug effects , Molecular Docking Simulation , Structure-Activity Relationship , Toremifene/chemistry , Toremifene/metabolism , Toremifene/pharmacology , Vero Cells , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism
7.
Medicine (Baltimore) ; 99(2): e18550, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31914031

ABSTRACT

BACKGROUND: Adjuvant endocrine therapy is a vital portion of postoperative comprehensive treatment for breast cancer patients. In recent years, studies have shown that endocrine therapy has a certain impact on the serum lipids of breast cancer patients, and the changes of lipid profiles may bring a series of problems. However, very few studies focus on this issue to date. The results of these studies are inconsistent, and the influence of different adjuvant endocrine modalities on lipid profiles still remains controversial. In order to better explore this issue, we conduct this network meta-analysis. METHOD: The protocol followed preferred reporting items for systematic reviews and meta-analyses protocols. Three main databases (PubMed, Embase, and the Cochrane Library) will be searched systematically for eligible randomized controlled trials without language restriction. In addition, a manual search of the references of relevant published studies will also be considered. Two reviewers will conduct studies selection, data extraction, and risk of bias assessment independently. The primary outcome is the variation of biochemical parameters - the serum lipid profiles (cholesterol, triglyceride, high-density lipoprotein, low low-density lipoprotein). RESULTS: The results will provide useful information about the side effects of different adjuvant endocrine drugs on lipid profiles in postoperative breast cancer patients (estrogen receptor-positive and/or progesterone receptor-positive). CONCLUSION: The findings of this study will be published in a peer-reviewed journal. PROSPERO REGISTRATION NUMBER: CRD42019129850.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Lipids/blood , Anastrozole/pharmacology , Anastrozole/therapeutic use , Androstadienes/pharmacology , Androstadienes/therapeutic use , Aromatase Inhibitors/pharmacology , Aromatase Inhibitors/therapeutic use , Chemotherapy, Adjuvant , Humans , Letrozole/pharmacology , Letrozole/therapeutic use , Network Meta-Analysis , Research Design , Tamoxifen/pharmacology , Tamoxifen/therapeutic use , Toremifene/pharmacology , Toremifene/therapeutic use
8.
Drug Res (Stuttg) ; 69(12): 683-687, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31550733

ABSTRACT

HER2-positive breast tumors are found in 25-30% of patients with breast cancer and are characterized by aggressive course and reduced sensitivity to both chemotherapy and hormone therapy. The aim of the work was to study the possibilities of enhancing the therapeutic effect of anti-estrogen drug toremifene by combining it with biguanide, metformin, on the HER2-positive breast cancer model in FVB/N HER-2/neu transgenic mouse. Male FBV/N mice with intramuscularly transplanted HER2-positive mammary gland tumor from a female mouse of the same strain have been given toremifene (30 mg/kg, orally daily) or metformin (100 mg/kg, orally daily) that had a moderate antitumor effect (decrease the area under the kinetic curve of tumor growth by 1.6 and 1.5 times, respectively, when compared with intact control). Co-administration of these drugs in the same doses had a more pronounced effect (the area under the kinetic curve of tumor growth decreased by 3.1 times compared to intact control; p<0.05). After 10 days, in group receiving toremifene all 10 mice developed inguinal-scrotal hernias, and in group that received toremifene plus metformin - only 5 of 10 (p=0.0325). By the 15th day after the start of treatment, the hernias was also determined in all mice treated with the combination of toremifene and metformin, but the size of the hernial sac was significantly smaller than in those receiving only toremifene - 537 ± 96 mm3 and 1309 ± 120 mm3, respectively (p=0.0001). A possible explanation is the manifestation of collagen-degrading effect of toremifene.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms, Male/drug therapy , Breast Neoplasms/drug therapy , Metformin/pharmacology , Receptor, ErbB-2/metabolism , Toremifene/pharmacology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms, Male/metabolism , Disease Models, Animal , Female , Male , Mice , Mice, Transgenic
9.
Lancet Infect Dis ; 19(9): 1023-1032, 2019 09.
Article in English | MEDLINE | ID: mdl-31300330

ABSTRACT

BACKGROUND: The ongoing Ebola virus outbreak in the Ituri and North Kivu Provinces of the Democratic Republic of the Congo, which began in July, 2018, is the second largest ever recorded. Despite civil unrest, outbreak control measures and the administration of experimental therapies and a vaccine have been initiated. The aim of this study was to test the efficacy of candidate therapies and diagnostic tests with the outbreak strain Ituri Ebola virus. Lacking a virus isolate from this outbreak, a recombinant Ituri Ebola virus was compared with a similarly engineered Makona virus from the 2013-16 outbreak. METHODS: Using Ebola virus sequences provided by organisations in DR Congo and a reverse genetics system, we generated an authentic Ebola virus from the ongoing outbreak in Ituri and North Kivu provinces. To relate this virus to other Ebola viruses in DR Congo, we did a phylogenetic analysis of representative complete Ebola virus genome sequences from previous outbreaks. We evaluated experimental therapies being tested in clinical trials in DR Congo, including remdesivir and ZMapp monoclonal antibodies, for their ability to inhibit the growth of infectious Ituri Ebola virus in cell culture. We also tested diagnostic assays for detection of the Ituri Ebola virus sequence. FINDINGS: The phylogenetic analysis of whole-genome sequences from each Ebola virus outbreak suggests there are at least two Ebola virus strains in DR Congo, which have independently crossed into the human population. The Ituri Ebola strain initially grew slower than the Makona strain, yet reached similar mean yields of 3 × 107 50% tissue culture infectious dose by 72 h infection in Huh-7 cells. Ituri Ebola virus was similar to Makona in its susceptibility to inhibition by remdesivir and to neutralisation by monoclonal antibodies from ZMapp and other monoclonal antibodies. Remdesivir inhibited Ituri Ebola virus at a 50% effective concentration (EC50) of 12nM (with a selectivity index of 303) and Makona Ebola virus at 13nM (with a selectivity index of 279). The Zmapp monoclonal antibodies 2G4 and 4G7 neutralised Ituri Ebola virus with a mean EC50 of 0·24 µg/mL and 0·48 µg/mL, and Makona Ebola virus with a mean EC50 of 0·45 µg/mL and 0·2 µg/mL. The Xpert Ebola and US Centers for Disease Control and Prevention real-time RT-qPCR diagnostic assays detected Ituri and Makona Ebola virus sequences with similar sensitivities and efficiencies, despite primer site binding mismatches in the Ituri Ebola virus. INTERPRETATION: Our findings provide a rationale for the continued testing of investigational therapies, confirm the effectiveness of the diagnostic assays used in the region, and establish a paradigm for the use of reverse genetics to inform response activities in an outbreak. FUNDING: US Centers for Disease Control and Prevention.


Subject(s)
Antiviral Agents/pharmacology , DNA, Viral/analysis , Disease Outbreaks , Ebolavirus/drug effects , Ebolavirus/genetics , Hemorrhagic Fever, Ebola/epidemiology , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Amiodarone/pharmacology , Anti-Arrhythmia Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Calcium Channel Blockers/pharmacology , Cell Line , Democratic Republic of the Congo/epidemiology , Humans , Phylogeny , Ribavirin/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Toremifene/pharmacology , Verapamil/pharmacology , Virus Cultivation , Whole Genome Sequencing
10.
Int J Hyg Environ Health ; 222(4): 670-677, 2019 05.
Article in English | MEDLINE | ID: mdl-31129136

ABSTRACT

Endocrine active substances (EAS), which are commonly used in pharmaceuticals and personal care products, are released into surface water mainly through WWTP effluents and have been shown to cause adverse effects in aquatic organisms. In wastewater, a variety of EAS with different hormonal activities is present, which can lead to additive effects or mask an endocrine activity. To investigate hormonal combination effects, with a focus on estrogen and androgen-modulators, influent samples from municipal and hospital wastewater treatmenr plants were spiked with 17α-ethinylestradiol, toremifene, 17α-methyltestosterone and bicalutamide and analyzed using in vitro reporter gene CALUX assays. All wastewaters caused endocrine activities in human cells, which were modified by adding one or several endocrine active substances. As expected, estrogenic activity was reduced in presence of the anti-estrogenic toremifene and androgenic activity decreased with the anti-androgen bicalutamide. In general, substance addition caused a similar trend in altered endocrine activities; however, their intensities differed between the wastewaters. Our results indicate that masking effects, leading to a suppressed biological signal, are of significant importance in the assessment of complex water samples, and combination effects rather than single substances determine the final biological effect. This emphasizes the need of effect-based tools in the assessment of water samples.


Subject(s)
Androgens/pharmacology , Endocrine Disruptors/pharmacology , Estrogens/pharmacology , Wastewater , Water Pollutants, Chemical/pharmacology , Anilides/pharmacology , Biological Assay , Cell Line, Tumor , Drug Interactions , Ethinyl Estradiol/pharmacology , Genes, Reporter , Humans , Luciferases/genetics , Methyltestosterone/pharmacology , Nitriles/pharmacology , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Toremifene/pharmacology , Tosyl Compounds/pharmacology
11.
Analyst ; 144(8): 2725-2735, 2019 Apr 08.
Article in English | MEDLINE | ID: mdl-30865733

ABSTRACT

Fourier transform infrared spectroscopy (FTIR) has been shown to be a promising tool for identifying the mode of action of drugs. However, most previous studies have focused on the analysis of fixed or dried cells. The measurement of living cells has the advantage of obtaining time series data, and the in situ approach eliminates the need for fixing or drying the cells. In this study, the potential of live-cell FTIR method for the identification of the mode of action of drugs was demonstrated. Four different drugs were tested, with two of the drugs having the same mode of action (tamoxifen and toremifene) and the other two having different modes of action (imatinib and doxorubicin). Live cells were treated in the four drugs at and below the IC50 level (i.e. the concentration of drug required to inhibit the growth of cells by 50%), and the changes to their spectra after the addition of drugs were monitored over a 24-hour period. Principal component analysis (PCA) of the spectral data shows that drugs with different modes of action are well-separated, while the drugs with the same mode of action are grouped together. The results also show that at IC50, the separation appears to be the clearest at 2 hours for imatinib and tamoxifen/toremifene and 6 hours for doxorubicin. However, at 50% of the IC50 drug concentration, the separation appears to be the best at longer incubation time, i.e. 24 hours, for all four drugs. In conclusion, live-cell FTIR has shown to be able to distinguish and group spectral signatures of cells treated with drugs of known modes of action after a relatively short time of exposure. Further collection of live-cell data would enable an algorithm to be developed for the prediction of the modes of action of novel drugs, which can help in the preclinical drug screening process.


Subject(s)
Antineoplastic Agents/classification , Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Imatinib Mesylate/pharmacology , Tamoxifen/pharmacology , Toremifene/pharmacology , Cell Line, Tumor , Humans , Inhibitory Concentration 50 , Principal Component Analysis , Spectroscopy, Fourier Transform Infrared/methods
12.
mBio ; 9(6)2018 12 18.
Article in English | MEDLINE | ID: mdl-30563895

ABSTRACT

The concept of repurposing previously approved medications to the treatment of new indications by taking advantage of off-target effects has gained traction in recent years, particularly in areas of medicine that do not offer large profits to pharmaceutical firms. As infectious disease discovery research has declined among large pharmaceutical companies, the potential payoff of repurposing has become attractive. From these efforts, the triphenylethylene class of selective estrogen receptor modulators related to tamoxifen has shown activity against a wide range of medically important human pathogens, including bacteria, fungi, parasites, and viruses. Because it has activity against many pathogens affecting people in resource-limited areas of the world, TAM and related drugs may be particularly useful. Here, we review the in vitro, in vivo, and mechanistic studies of the anti-infective activity of tamoxifen, toremifene, clomiphene, and their analogs. We also discuss the pharmacologic properties of this privileged scaffold and its potential utility in treating infectious diseases.


Subject(s)
Communicable Diseases/drug therapy , Drug Repositioning , Estrogen Receptor Antagonists/pharmacology , Animals , Bacteria/drug effects , Female , Humans , Mice , Parasites/drug effects , Stilbenes/therapeutic use , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Toremifene/analogs & derivatives , Toremifene/pharmacology , Viruses/drug effects
13.
PLoS Negl Trop Dis ; 11(4): e0005540, 2017 04.
Article in English | MEDLINE | ID: mdl-28403145

ABSTRACT

Phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) is a lipid kinase involved in endosome maturation that emerged from a haploid genetic screen as being required for Ebola virus (EBOV) infection. Here we analyzed the effects of apilimod, a PIKfyve inhibitor that was reported to be well tolerated in humans in phase 2 clinical trials, for its effects on entry and infection of EBOV and Marburg virus (MARV). We first found that apilimod blocks infections by EBOV and MARV in Huh 7, Vero E6 and primary human macrophage cells, with notable potency in the macrophages (IC50, 10 nM). We next observed that similar doses of apilimod block EBOV-glycoprotein-virus like particle (VLP) entry and transcription-replication competent VLP infection, suggesting that the primary mode of action of apilimod is as an entry inhibitor, preventing release of the viral genome into the cytoplasm to initiate replication. After providing evidence that the anti-EBOV action of apilimod is via PIKfyve, we showed that it blocks trafficking of EBOV VLPs to endolysosomes containing Niemann-Pick C1 (NPC1), the intracellular receptor for EBOV. Concurrently apilimod caused VLPs to accumulate in early endosome antigen 1-positive endosomes. We did not detect any effects of apilimod on bulk endosome acidification, on the activity of cathepsins B and L, or on cholesterol export from endolysosomes. Hence by antagonizing PIKfyve, apilimod appears to block EBOV trafficking to its site of fusion and entry into the cytoplasm. Given the drug's observed anti-filoviral activity, relatively unexplored mechanism of entry inhibition, and reported tolerability in humans, we propose that apilimod be further explored as part of a therapeutic regimen to treat filoviral infections.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Marburgvirus/drug effects , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Triazines/pharmacology , Virus Internalization/drug effects , Animals , Biological Transport , Cell Line , Chlorocebus aethiops , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/drug therapy , Humans , Hydrazones , Lysosomes/metabolism , Macrophages/virology , Marburgvirus/physiology , Nocodazole/pharmacology , Pyrimidines , Toremifene/pharmacology , Vero Cells
14.
Article in English | MEDLINE | ID: mdl-27993858

ABSTRACT

The spread of antibiotic resistance and the challenges associated with antiseptics such as chlorhexidine have necessitated a search for new antibacterial agents against oral bacterial pathogens. As a result of failing traditional approaches, drug repurposing has emerged as a novel paradigm to find new antibacterial agents. In this study, we examined the effects of the FDA-approved anticancer agent toremifene against the oral bacteria Porphyromonas gingivalis and Streptococcus mutans We found that the drug was able to inhibit the growth of both pathogens, as well as prevent biofilm formation, at concentrations ranging from 12.5 to 25 µM. Moreover, toremifene was shown to eradicate preformed biofilms at concentrations ranging from 25 to 50 µM. In addition, we found that toremifene prevents P. gingivalis and S. mutans biofilm formation on titanium surfaces. A time-kill study indicated that toremifene is bactericidal against S. mutans Macromolecular synthesis assays revealed that treatment with toremifene does not cause preferential inhibition of DNA, RNA, or protein synthesis pathways, indicating membrane-damaging activity. Biophysical studies using fluorescent probes and fluorescence microscopy further confirmed the membrane-damaging mode of action. Taken together, our results suggest that the anticancer agent toremifene is a suitable candidate for further investigation for the development of new treatment strategies for oral bacterial infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Biofilms/drug effects , Cell Membrane/drug effects , Porphyromonas gingivalis/drug effects , Streptococcus mutans/drug effects , Toremifene/pharmacology , Biofilms/growth & development , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Membrane Permeability/drug effects , Dental Plaque/drug therapy , Dental Plaque/microbiology , Drug Repositioning , Drug Resistance, Multiple, Bacterial/physiology , Humans , Microbial Sensitivity Tests , Periodontitis/drug therapy , Periodontitis/microbiology , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/ultrastructure , Streptococcus mutans/metabolism , Streptococcus mutans/ultrastructure , Titanium/analysis
15.
Antiviral Res ; 137: 165-172, 2017 01.
Article in English | MEDLINE | ID: mdl-27890675

ABSTRACT

Although a group of FDA-approved drugs were previously identified with activity against Ebola virus (EBOV), most of them are not clinically useful because their human blood concentrations are not high enough to inhibit EBOV infection. We screened 795 unique three-drug combinations in an EBOV entry assay. Two sets of three-drug combinations, toremifene-mefloquine-posaconazole and toremifene-clarithromycin-posaconazole, were identified that effectively blocked EBOV entry and were further validated for inhibition of live EBOV infection. The individual drug concentrations in the combinations were reduced to clinically relevant levels. We identified mechanisms of action of these drugs: functional inhibitions of Niemann-Pick C1, acid sphingomyelinase, and lysosomal calcium release. Our findings identify the drug combinations with potential to treat EBOV infection.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Virus Internalization/drug effects , Animals , Cell Line , Chlorocebus aethiops , Clarithromycin/pharmacology , Drug Combinations , Drug Synergism , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , High-Throughput Screening Assays , Humans , Mefloquine/pharmacology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/drug effects , Toremifene/pharmacology , Triazoles/pharmacology , Vero Cells
16.
Microb Drug Resist ; 23(5): 640-650, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27935770

ABSTRACT

Novel therapeutic approaches are urgently needed to combat nosocomial infections caused by extremely drug-resistant (XDR) "superbugs." This study aimed to investigate the synergistic antibacterial activity of polymyxin B in combination with selective estrogen receptor modulators (SERMs) against problematic Gram-negative pathogens. In vitro synergistic antibacterial activity of polymyxin B and the SERMs tamoxifen, raloxifene, and toremifene was assessed using the microdilution checkerboard and static time-kill assays against a panel of Gram-negative isolates. Polymyxin B and the SERMs were ineffective when used as monotherapy against polymyxin-resistant minimum inhibitory concentration ([MIC] ≥8 mg/L) Pseudomonas aeruginosa, Klebsiella pneumoniae, and Acinetobacter baumannii. However, when used in combination, clinically relevant concentrations of polymyxin B and SERMs displayed synergistic killing against the polymyxin-resistant P. aeruginosa, K. pneumoniae, and A. baumannii isolates as demonstrated by a ≥2-3 log10 decrease in bacterial count (CFU/ml) after 24 hours. The combination of polymyxin B with toremifene demonstrated very potent antibacterial activity against P. aeruginosa biofilms in an artificial sputum media assay. Moreover, polymyxin B combined with toremifene synergistically induced cytosolic green fluorescence protein release, cytoplasmic membrane depolarization, permeabilizing activity in a nitrocefin assay, and an increase of cellular reactive oxygen species from P. aeruginosa cells. In addition, scanning and transmission electron micrographs showed that polymyxin B in combination with toremifene causes distinctive damage to the outer membrane of P. aeruginosa cells, compared with treatments with each compound per se. In conclusion, the combination of polymyxin B and SERMs illustrated a synergistic activity against XDR Gram-negative pathogens, including highly polymyxin-resistant P. aeruginosa isolates, and represents a novel combination therapy strategy for the treatment of infections because of problematic XDR Gram-negative pathogens.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Polymyxin B/pharmacology , Raloxifene Hydrochloride/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Tamoxifen/pharmacology , Toremifene/pharmacology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/growth & development , Acinetobacter baumannii/ultrastructure , Biofilms/growth & development , Cell Membrane Permeability/drug effects , Drug Repositioning , Drug Resistance, Multiple, Bacterial/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Drug Synergism , Drug Therapy, Combination , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/growth & development , Klebsiella pneumoniae/ultrastructure , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/ultrastructure
17.
Sci Rep ; 6: 32105, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27601313

ABSTRACT

Selective estrogen receptor modulators (SERMs) are widely prescribed drugs that alter cellular and whole-body cholesterol homeostasis. Here we evaluate the effect of SERMs on the macrophage-specific reverse cholesterol transport (M-RCT) pathway, which is mediated by HDL. Treatment of human and mouse macrophages with tamoxifen, raloxifene or toremifene induced the accumulation of cytoplasmic vesicles of acetyl-LDL-derived free cholesterol. The SERMs impaired cholesterol efflux to apolipoprotein A-I and HDL, and lowered ABCA1 and ABCG1 expression. These effects were not altered by the antiestrogen ICI 182,780 nor were they reproduced by 17ß-estradiol. The treatment of mice with tamoxifen or raloxifene accelerated HDL-cholesteryl ester catabolism, thereby reducing HDL-cholesterol concentrations in serum. When [(3)H]cholesterol-loaded macrophages were injected into mice intraperitoneally, tamoxifen, but not raloxifene, decreased the [(3)H]cholesterol levels in serum, liver and feces. Both SERMs downregulated liver ABCG5 and ABCG8 protein expression, but tamoxifen reduced the capacity of HDL and plasma to promote macrophage cholesterol efflux to a greater extent than raloxifene. We conclude that SERMs interfere with intracellular cholesterol trafficking and efflux from macrophages. Tamoxifen, but not raloxifene, impair M-RCT in vivo. This effect is primarily attributable to the tamoxifen-mediated reduction of the capacity of HDL to promote cholesterol mobilization from macrophages.


Subject(s)
Cholesterol/metabolism , Lipoproteins, HDL/metabolism , Macrophages/drug effects , Selective Estrogen Receptor Modulators/pharmacology , ATP Binding Cassette Transporter 1/biosynthesis , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter, Subfamily G/biosynthesis , ATP Binding Cassette Transporter, Subfamily G/genetics , Acetyl-CoA C-Acetyltransferase/antagonists & inhibitors , Animals , Apolipoprotein A-I/metabolism , Biological Transport/drug effects , Cholesterol/analysis , Cholesterol/blood , Cholesterol Esters/metabolism , Diet, Western , Esterification/drug effects , Estradiol/analogs & derivatives , Estradiol/pharmacology , Feces/chemistry , Fulvestrant , Humans , Lipoproteins, LDL/metabolism , Liver/chemistry , Liver/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Raloxifene Hydrochloride/pharmacology , THP-1 Cells , Tamoxifen/pharmacology , Toremifene/pharmacology
18.
Nature ; 535(7610): 169-172, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27362232

ABSTRACT

Ebola viruses (EBOVs) are responsible for repeated outbreaks of fatal infections, including the recent deadly epidemic in West Africa. There are currently no approved therapeutic drugs or vaccines for the disease. EBOV has a membrane envelope decorated by trimers of a glycoprotein (GP, cleaved by furin to form GP1 and GP2 subunits), which is solely responsible for host cell attachment, endosomal entry and membrane fusion. GP is thus a primary target for the development of antiviral drugs. Here we report the first, to our knowledge, unliganded structure of EBOV GP, and high-resolution complexes of GP with the anticancer drug toremifene and the painkiller ibuprofen. The high-resolution apo structure gives a more complete and accurate picture of the molecule, and allows conformational changes introduced by antibody and receptor binding to be deciphered. Unexpectedly, both toremifene and ibuprofen bind in a cavity between the attachment (GP1) and fusion (GP2) subunits at the entrance to a large tunnel that links with equivalent tunnels from the other monomers of the trimer at the three-fold axis. Protein­drug interactions with both GP1 and GP2 are predominately hydrophobic. Residues lining the binding site are highly conserved among filoviruses except Marburg virus (MARV), suggesting that MARV may not bind these drugs. Thermal shift assays show up to a 14 °C decrease in the protein melting temperature after toremifene binding, while ibuprofen has only a marginal effect and is a less potent inhibitor. These results suggest that inhibitor binding destabilizes GP and triggers premature release of GP2, thereby preventing fusion between the viral and endosome membranes. Thus, these complex structures reveal the mechanism of inhibition and may guide the development of more powerful anti-EBOV drugs.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/metabolism , Ebolavirus/chemistry , Toremifene/chemistry , Toremifene/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antiviral Agents/pharmacology , Binding Sites , Cell Line , Conserved Sequence , Ebolavirus/drug effects , Endosomes/drug effects , Endosomes/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Ibuprofen/chemistry , Ibuprofen/metabolism , Ibuprofen/pharmacology , Ligands , Marburgvirus/chemistry , Membrane Fusion/drug effects , Models, Molecular , Protein Binding , Protein Stability/drug effects , Protein Structure, Quaternary/drug effects , Protein Subunits/chemistry , Protein Subunits/metabolism , Temperature , Toremifene/pharmacology , Viral Envelope Proteins/antagonists & inhibitors , Virus Attachment/drug effects
19.
PLoS Negl Trop Dis ; 10(1): e0004364, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26752302

ABSTRACT

To date there are no approved antiviral drugs for the treatment of Ebola virus disease (EVD). While a number of candidate drugs have shown limited efficacy in vitro and/or in non-human primate studies, differences in experimental methodologies make it difficult to compare their therapeutic effectiveness. Using an in vitro model of Ebola Zaire replication with transcription-competent virus like particles (trVLPs), requiring only level 2 biosafety containment, we compared the activities of the type I interferons (IFNs) IFN-α and IFN-ß, a panel of viral polymerase inhibitors (lamivudine (3TC), zidovudine (AZT) tenofovir (TFV), favipiravir (FPV), the active metabolite of brincidofovir, cidofovir (CDF)), and the estrogen receptor modulator, toremifene (TOR), in inhibiting viral replication in dose-response and time course studies. We also tested 28 two- and 56 three-drug combinations against Ebola replication. IFN-α and IFN-ß inhibited viral replication 24 hours post-infection (IC50 0.038µM and 0.016µM, respectively). 3TC, AZT and TFV inhibited Ebola replication when used alone (50-62%) or in combination (87%). They exhibited lower IC50 (0.98-6.2µM) compared with FPV (36.8µM), when administered 24 hours post-infection. Unexpectedly, CDF had a narrow therapeutic window (6.25-25µM). When dosed >50µM, CDF treatment enhanced viral infection. IFN-ß exhibited strong synergy with 3TC (97.3% inhibition) or in triple combination with 3TC and AZT (95.8% inhibition). This study demonstrates that IFNs and viral polymerase inhibitors may have utility in EVD. We identified several 2 and 3 drug combinations with strong anti-Ebola activity, confirmed in studies using fully infectious ZEBOV, providing a rationale for testing combination therapies in animal models of lethal Ebola challenge. These studies open up new possibilities for novel therapeutic options, in particular combination therapies, which could prevent and treat Ebola infection and potentially reduce drug resistance.


Subject(s)
Ebolavirus/drug effects , Interferon-beta/pharmacology , Nucleosides/therapeutic use , Virus Replication/drug effects , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacology , CCR5 Receptor Antagonists/administration & dosage , CCR5 Receptor Antagonists/pharmacology , Cyclohexanes/administration & dosage , Cyclohexanes/pharmacology , Humans , Maraviroc , Toremifene/administration & dosage , Toremifene/pharmacology , Triazoles/administration & dosage , Triazoles/pharmacology
20.
Post Reprod Health ; 21(3): 112-21, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26289836

ABSTRACT

Traditional menopausal hormone therapy containing estrogens/progestin has been associated with an increased risk of breast cancer, and estrogen exposure is known to promote growth and proliferation of a majority of breast cancers. Therefore, it is important for clinicians to consider the breast safety profile of any hormone-based therapy used in postmenopausal women. This review provides an overview of the breast safety and tolerability profiles of currently marketed selective estrogen receptor modulators, antiestrogens, and the first tissue selective estrogen complex combining conjugated estrogens with the selective estrogen receptor modulator bazedoxifene in postmenopausal women. Selective estrogen receptor modulators and antiestrogens act as estrogen receptor antagonists in the breast. Tamoxifen, toremifene, and the selective estrogen receptor degrader fulvestrant are used to treat breast cancer, and tamoxifen and raloxifene protect against breast cancer in high-risk women. Postmenopausal women using selective estrogen receptor modulators for prevention or treatment of osteoporosis (raloxifene, bazedoxifene) can be reassured that these hormonal treatments do not adversely affect their risk of breast cancer and may, in the case of raloxifene, even be protective. There are limited data on breast cancer in women who use ospemifene for dyspareunia. Conjugated estrogens/bazedoxifene use for up to two years did not increase mammographic breast density or breast pain/tenderness, and there was no evidence of an increased risk of breast cancer, suggesting that conjugated estrogens/bazedoxifene has an improved breast safety profile compared with traditional menopausal hormone therapies. Future research will continue to focus on development of selective estrogen receptor modulators and selective estrogen receptor modulator combinations capable of achieving the ideal balance of estrogen receptor agonist and antagonist effects.


Subject(s)
Breast Neoplasms/epidemiology , Breast/drug effects , Estrogen Antagonists/pharmacology , Estrogens, Conjugated (USP)/pharmacology , Estrogens/pharmacology , Indoles/pharmacology , Selective Estrogen Receptor Modulators/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/prevention & control , Drug Therapy, Combination , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Antagonists/therapeutic use , Estrogens/therapeutic use , Estrogens, Conjugated (USP)/therapeutic use , Female , Fulvestrant , Humans , Indoles/therapeutic use , Osteoporosis/prevention & control , Postmenopause , Protective Factors , Raloxifene Hydrochloride/pharmacology , Risk Assessment , Selective Estrogen Receptor Modulators/therapeutic use , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Toremifene/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...