Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Anal Chem ; 96(8): 3454-3461, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38359782

ABSTRACT

Estrogen receptor α (ERα) is an important biomarker in breast cancer diagnosis and treatment. Sensitive and accurate detection of ERα protein expression is crucial in guiding selection of an appropriate therapeutic strategy to improve the effectiveness and prognosis of breast cancer treatment. Herein, we report a liquid-gated graphene field-effect transistor (FET) biosensor that enables rapid, sensitive, and label-free detection of the ERα protein by employing a novel drug molecule as a capture probe. The drug molecule was synthesized and subsequently immobilized onto the sensing surface of the fabricated graphene FET, which was able to distinguish the ERα-positive from the ERα-negative protein. The developed sensor not only demonstrated a low detection limit (LOD: 2.62 fM) but also achieved a fast response to ERα protein samples within 30 min. Moreover, depending on the relationship between the change of dirac point and the ERα protein concentrations, the dissociation constant (Kd) was estimated to be 7.35 ± 0.06 pM, indicating that the drug probe-modified graphene FET had a good affinity with ERα protein. The nanosensor was able to analyze ERα proteins from 36 cell samples lysates. These results show that the graphene FET sensor was able to differentiate between ERα-positive and ERα-negative cells, indicating a promising biosensor for the ultrasensitive and rapid detection of ERα protein without antibody labeling.


Subject(s)
Biosensing Techniques , Graphite , Limit of Detection , Estrogen Receptor alpha , Transistors, Electronic , Biomarkers , Biosensing Techniques/methods
2.
Chembiochem ; 24(4): e202200680, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36564338

ABSTRACT

As an emerging therapeutic strategy, proteolysis-targeting chimeras (PROTACs) have been proven to be superior to traditional drugs in many aspects. However, due to their unique mechanism of action, existing methods for evaluating the degradation still have many limitations, which seriously restricts the development of PROTACs. In this methodological study, using direct stochastic optical reconstruction microscopy (dSTORM)-based single-cell protein quantitative analysis, we systematically investigated the dynamic degradation characteristics of FLT3 protein during PROTACs treatment. We found that the distribution of FLT3 varies between FLT3-ITD mutation and FLT3-WT cells. PROTACs had an obvious time-course effect on protein degradation and present two distinct phases; this provided a basis for deciding when to evaluate protein degradation. High concentrations of PROTACs were more effective than long-time administration because a higher Dmax was achieved. Two-color dSTORM-based colocalization analysis efficiently detected the proportion of ternary complexes, making it very useful in screening PROTACs. Taken together, our findings show that the dSTORM method is an ideal tool for evaluating PROTACs and will accelerate the development of new PROTACs.


Subject(s)
Microscopy , Proteins , Proteins/metabolism , Proteolysis
3.
Bioorg Med Chem ; 82: 117235, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36905762

ABSTRACT

Breast cancer (BC), a well-known estrogen-dependent cancer, is the most common cancer among women and the leading cause of cancer deaths. One of the most important therapeutic approaches for BC is endocrine therapy targeting estrogen receptor alpha (ERα) and thus blocking the estrogen receptor signaling pathway. Drugs, such as tamoxifen or fulvestrant, are developed based on this theory and have benefited numerous patients with BC for many years. However, many patients with advanced BC, such as tamoxifen-resistant BC, cannot benefit from these developed drugs anymore. Therefore, new drugs targeting ERα are urgently needed by patients with BC. Recently, elacestrant, a novel selective estrogen receptor degrader (SERD), was approved by the United States Food and Drug Administration (FDA), highlighting the importance of ERα degradation in endocrine therapy. Proteolysis targeting chimera (PROTAC) has been considered a powerful technique for targeting protein degradation (TPD). In this regard, we developed and studied a novel ERα degrader, which is a PROTAC-like SERD named 17e. We found that compound 17e can inhibit the growth of BC both in vitro and in vivo and induce the cell cycle arrest of BC. Importantly, 17e displayed no apparent toxicity toward healthy kidney and liver cells. Moreover, we observed that the presence of 17e led to a dramatic increase in the autophagy-lysosome pathway in an ERα-independent manner. Finally, we revealed that a decrease in MYC, a frequent deregulation oncogene in human cancers, was mediated by both ERα degradation and autophagy activation in the presence of 17e. Collectively, we discovered that compound 17e induced ERα degradation and exerts significant anti-cancer effects on BC mainly through promoting the autophagy-lysosome pathway and decreasing MYC level.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Receptors, Estrogen/metabolism , Cell Proliferation , Estrogen Antagonists/pharmacology , Tamoxifen/pharmacology , Cell Cycle Checkpoints , MCF-7 Cells , Cell Line, Tumor
4.
Int J Mol Sci ; 24(8)2023 Apr 12.
Article in English | MEDLINE | ID: mdl-37108300

ABSTRACT

Due to the poor metabolic conditions fomenting the emergence of the Warburg effect (WE) phenotype, abnormal glycometabolism has become a unique and fundamental research topic in the field of tumor biology. Moreover, hyperglycemia and hyperinsulinism are associated with poor outcomes in patients with breast cancer. However, there are a few studies on anticancer drugs targeting glycometabolism in breast cancer. We hypothesized that Oxabicycloheptene sulfonate (OBHS), a class of compounds that function as selective estrogen receptor modulators, may hold potential in a therapy for breast cancer glycometabolism. Here, we evaluated concentrations of glucose, glucose transporters, lactate, 40 metabolic intermediates, and glycolytic enzymes using an enzyme-linked immunosorbent assay, Western blotting, and targeted metabolomic analysis in, in vitro and in vivo breast cancer models. OBHS significantly inhibited the expression of glucose transporter 1 (GLUT1) via PI3K/Akt signaling pathway to suppress breast cancer progression and proliferation. Following an investigation of the modulatory effect of OBHS on breast cancer cells, we found that OBHS suppressed the glucose phosphorylation and oxidative phosphorylation of glycolytic enzymes, leading to the decreased biological synthesis of ATP. This study was novel in highlighting the role of OBHS in the remodeling of tumor glycometabolism in breast cancer, and this is worth further investigation of breast cancer in clinical trials.


Subject(s)
Neoplasms , Phosphatidylinositol 3-Kinases , Humans , Glucose Transporter Type 1/genetics , Phosphatidylinositol 3-Kinases/metabolism , Neoplasms/metabolism , Glucose/metabolism , Cell Line, Tumor
5.
Molecules ; 28(5)2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36903555

ABSTRACT

Hydrogen peroxide is one of the most important reactive oxygen species, which plays a vital role in many physiological and pathological processes. A dramatic increase in H2O2 levels is a prominent feature of cancer. Therefore, rapid and sensitive detection of H2O2 in vivo is quite conducive to an early cancer diagnosis. On the other hand, the therapeutic potential of estrogen receptor beta (ERß) has been implicated in many diseases including prostate cancer, and this target has attracted intensive attention recently. In this work, we report the development of the first H2O2-triggered ERß-targeted near-infrared fluorescence (NIR) probe and its application in imaging of prostate cancer both in vitro and in vivo. The probe showed good ERß selective binding affinity, excellent H2O2 responsiveness and near infrared imaging potential. Moreover, in vivo and ex vivo imaging studies indicated that the probe could selectively bind to DU-145 prostate cancer cells and rapidly visualizes H2O2 in DU-145 xenograft tumors. Mechanistic studies such as high-resolution mass spectrometry (HRMS) and density functional theory (DFT) calculations indicated that the borate ester group is vital for the H2O2 response turn-on fluorescence of the probe. Therefore, this probe might be a promising imaging tool for monitoring the H2O2 levels and early diagnosis studies in prostate cancer research.


Subject(s)
Hydrogen Peroxide , Prostatic Neoplasms , Humans , Male , Diagnostic Imaging , Estrogen Receptor beta , Fluorescence , Fluorescent Dyes/chemistry , Hydrogen Peroxide/chemistry , Animals
6.
Bioorg Chem ; 122: 105683, 2022 05.
Article in English | MEDLINE | ID: mdl-35278779

ABSTRACT

Enterovirus A71 (EV-A71), one of the major pathogens that causes hand, foot and mouth disease (HFMD), has seriously threatened the health and safety of young children. In this study, aminothiazole derivatives were synthesized and screened against EV-A71 in Rhabdomyosarcoma (RD) cells. The best compound (12s), with a biphenyl group, showed activity against EV-A71 (EC50: 0.27 µM) but also against a series of different human enteroviruses without significant cytotoxicity (CC50 > 56.2 µM). Mechanistic studies including time-of-drug-addition assays, viral entry assays and microscale thermophoresis (MST) experiments, showed that 12s binds to EV-A71 capsid and blocks the binding between the viral protein VP1 and the relevant human scavenger receptor class B member 2 (hSCARB2).


Subject(s)
Capsid Proteins , Enterovirus A, Human , Thiazoles , Capsid Proteins/antagonists & inhibitors , Enterovirus A, Human/drug effects , Enterovirus Infections/drug therapy , Humans , Thiazoles/pharmacology , Virus Internalization
7.
Biochem Biophys Res Commun ; 573: 69-75, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34388457

ABSTRACT

Breast cancer (BRCA) is one of the most threatening cancer types, especially among the female population. 70% of breast cancer are estrogen receptor α (ERα) positive and endocrine therapy is effective to decrease breast cancer risk. Autophagy, a highly conserved cellular recycling process, has been regarded to serve a protective role in BRCA. Autophagy-related gene 13 (Atg13) is participated in autophagy and is critical to autophagy initiation. Briefly, we observed that ERα, a well-known transcription factor that can promote breast cancer cell proliferation, expressed higher in breast cancer tissues. Moreover, ERα had a significant positive correlation with Atg13 and may be able to regulate the transcription of Atg13 via binding the promoter region of Atg13. Surprisingly, Oxabicycloheptene sulfonate (OBHS), the drug that we reported as a selective estrogen receptor modulator (SERM) before, may have the ability to decrease the expression of ERα and suppress the autophagy. In conclusion. We found that ERα could be involved in autophagy by binding the promoter of Atg13, and compound OBHS may be able to affect the viability of breast cancer cells by decreasing the expression of ERα and Atg13.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy-Related Proteins/antagonists & inhibitors , Breast Neoplasms/drug therapy , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Sulfonic Acids/pharmacology , Autophagy-Related Proteins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Drug Screening Assays, Antitumor , Estrogen Receptor alpha/metabolism , HEK293 Cells , Humans
8.
Bioorg Med Chem ; 40: 116185, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33965842

ABSTRACT

Hormone therapy targeting estrogen receptors is widely used clinically for the treatment of breast cancer, such as tamoxifen, but most of them are partial agonists, which can cause serious side effects after long-term use. The use of selective estrogen receptor down-regulators (SERDs) may be an effective alternative to breast cancer therapy by directly degrading ERα protein to shut down ERα signaling. However, the solely clinically used SERD fulvestrant, is low orally bioavailable and requires intravenous injection, which severely limits its clinical application. On the other hand, double- or multi-target conjugates, which are able to synergize antitumor activity by different pathways, thus may enhance therapeutic effect in comparison with single targeted therapy. In this study, we designed and synthesized a series of novel dual-functional conjugates targeting both ERα degradation and histone deacetylase inhibiton by combining a privileged SERD skeleton 7-oxabicyclo[2.2.1]heptane sulfonamide (OBHSA) with a histone deacetylase inhibitor side chain. We found that substituents on both the sulfonamide nitrogen and phenyl group of OBHSA unit had significant effect on biological activities. Among them, conjugate 16i with N-methyl and naphthyl groups exhibited potent antiproliferative activity against MCF-7 cells, and excellent ERα degradation activity and HDACs inhibitory ability. A further molecular docking study indicated the interaction patterns of these conjugates with ERα, which may provide guidance to design novel SERDs or PROTAC-like SERDs for breast cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Sulfonamides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Estrogen Antagonists/chemical synthesis , Estrogen Antagonists/chemistry , Estrogen Receptor alpha/metabolism , Female , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Humans , MCF-7 Cells , Molecular Structure , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
9.
Mol Cell Biochem ; 463(1-2): 161-173, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31595422

ABSTRACT

Alzheimer's disease (AD) is the leading cause of dementia, which characterized by toxic senile plaques is composed of amyloid-ß (Aß). ß-site amyloid precursor protein-cleaving enzyme 1 (BACE1) is the rate-limiting protease in Aß generation. Therefore, pharmacology BACE1 inhibition is one of the prime targets for potential treatment of AD. Curcumin, a yellow polyphenol derived from the rhizomes of the plant Curcuma longa Linn, has been reported to cross the blood-brain barrier and prevent Aß aggregation in AD models. However, its neuroprotective mechanism is still unclear. In the present study, we find that curcumin markedly reduces Aß levels in HEK293-APPswe cells. Our results show that curcumin inhibits BACE1 gene expression in SH-SY5Y cells at transcriptional and translational levels. Furthermore, we reveal that nuclear factor kappa B (NFκB) signaling is involved in the regulation of curcumin on BACE1. Interestingly, the estrogenicity of curcumin is found to partially contribute to its protective action. Our data show that curcumin activates estrogen receptor ß (ERß) selectively and the activation of ERß directly effects on the upstream factors of the NFκB signaling pathway. The above results indicate that curcumin reduces BACE1 expression through ERß and NFκB pathway, providing a novel mechanism for curcumin as a candidate for AD therapy.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Curcumin/pharmacology , Estrogen Receptor beta/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/genetics , Aspartic Acid Endopeptidases/genetics , Cell Line, Tumor , Estrogen Receptor beta/genetics , HEK293 Cells , Humans , NF-kappa B/genetics , Signal Transduction/genetics
10.
Analyst ; 145(18): 5989-5995, 2020 Sep 14.
Article in English | MEDLINE | ID: mdl-32856648

ABSTRACT

ERα-targeted fluorescent probes are important tools for ERα study. In order to develop high quality ERα-targeted probes, a sound and complete evaluation system is essential but has not been established yet. Herein, we set up a series of evaluation criteria for ERα-targeted fluorescent probes including ERα binding affinity, fluorescence quantum yield, cytotoxicity, ERα tracking capacity, ERα selectivity and ERα labeling ability. To verify the practicability of the evaluation criteria, we designed and synthesized two ERα-targeted fluorescent probes and fully characterized their properties based on the proposed evaluation criteria. It showed that the probes exhibited better performance. Moreover, we applied the probes in MCF-7 cells to study the ERα motion characteristics for the first time. We hope that our evaluation criteria could be helpful for the establishment of a complete evaluation system for ERα-targeted fluorescent probes.


Subject(s)
Estrogen Receptor alpha , Fluorescent Dyes , Estrogen Receptor alpha/genetics , Humans , MCF-7 Cells
11.
Nat Chem Biol ; 13(1): 111-118, 2017 01.
Article in English | MEDLINE | ID: mdl-27870835

ABSTRACT

Resistance to endocrine therapies remains a major clinical problem for the treatment of estrogen receptor-α (ERα)-positive breast cancer. On-target side effects limit therapeutic compliance and use for chemoprevention, highlighting an unmet need for new therapies. Here we present a full-antagonist ligand series lacking the prototypical ligand side chain that has been universally used to engender antagonism of ERα through poorly understood structural mechanisms. A series of crystal structures and phenotypic assays reveal a structure-based design strategy with separate design elements for antagonism and degradation of the receptor, and access to a structurally distinct space for further improvements in ligand design. Understanding structural rules that guide ligands to produce diverse ERα-mediated phenotypes has broad implications for the treatment of breast cancer and other estrogen-sensitive aspects of human health including bone homeostasis, energy metabolism, and autoimmunity.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Humans , Ligands , Models, Molecular , Molecular Structure , Receptors, Estrogen/metabolism , Structure-Activity Relationship
12.
Org Biomol Chem ; 16(12): 2150-2155, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29517094

ABSTRACT

A 1,4-diazabicyclo[2.2.2]octane (DABCO) catalyzed selenocyclization of olefinic amides was achieved under mild reaction conditions. The reaction formed various benzeneselenyl substituted isobenzofuran-1(3H)-imine derivatives in good yields. The product was determined using single-crystal X-ray analysis. For compound 2u, the relative stereochemistry was established on the basis of NOESY NMR studies.

13.
Org Biomol Chem ; 16(3): 472-479, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29265146

ABSTRACT

A series of optically active pyrano[3,2-c]chromenes have been synthesized through an asymmetric domino reaction of 4-hydroxy-2H-chromen-2-ones with malononitriles. The targeted molecules were obtained in excellent yields and enantioselectivities (up to 94% yield, 99% ee). The AChE inhibitory activity studies revealed that compounds 4n (IC50 = 21.3 µM) and 4p (IC50 = 19.2 µM) displayed potent acetylcholinesterase inhibition. In most cases, the S-enantiomers were superior to the corresponding R-enantiomers. Moreover, molecular modelling provides a practical method for understanding the enantioselective discrimination of AChE with these kinds of compounds.


Subject(s)
Acetylcholinesterase/metabolism , Benzopyrans/chemical synthesis , Benzopyrans/pharmacology , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/pharmacology , Acetylcholinesterase/chemistry , Benzopyrans/chemistry , Benzopyrans/metabolism , Catalysis , Chemistry Techniques, Synthetic , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/metabolism , Inhibitory Concentration 50 , Molecular Docking Simulation , Protein Conformation , Stereoisomerism , Structure-Activity Relationship
14.
Mol Syst Biol ; 12(4): 864, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27107013

ABSTRACT

Some estrogen receptor-α (ERα)-targeted breast cancer therapies such as tamoxifen have tissue-selective or cell-specific activities, while others have similar activities in different cell types. To identify biophysical determinants of cell-specific signaling and breast cancer cell proliferation, we synthesized 241 ERα ligands based on 19 chemical scaffolds, and compared ligand response using quantitative bioassays for canonical ERα activities and X-ray crystallography. Ligands that regulate the dynamics and stability of the coactivator-binding site in the C-terminal ligand-binding domain, called activation function-2 (AF-2), showed similar activity profiles in different cell types. Such ligands induced breast cancer cell proliferation in a manner that was predicted by the canonical recruitment of the coactivators NCOA1/2/3 and induction of the GREB1 proliferative gene. For some ligand series, a single inter-atomic distance in the ligand-binding domain predicted their proliferative effects. In contrast, the N-terminal coactivator-binding site, activation function-1 (AF-1), determined cell-specific signaling induced by ligands that used alternate mechanisms to control cell proliferation. Thus, incorporating systems structural analyses with quantitative chemical biology reveals how ligands can achieve distinct allosteric signaling outcomes through ERα.


Subject(s)
Estrogen Receptor alpha/chemistry , Estrogen Receptor alpha/metabolism , Signal Transduction/drug effects , Binding Sites , Cell Proliferation/drug effects , Estrogen Receptor alpha/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , MCF-7 Cells , Models, Molecular , Molecular Structure , Peptide Library , Protein Binding
15.
Neurochem Res ; 42(8): 2246-2256, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28374135

ABSTRACT

Oxabicycloheptene sulfonate (OBHS) is a novel bicyclic core selective estrogen receptor modulator (SERM) with estrogen receptor (ER) antagonistic-activity and anti-inflammatory activity. However, little is known about protective action of OBHS on neurodegenerative disorders. In the present study, OBHS demonstrated a remarkably protective effect against amyloid beta (Aß) induced cytotoxicity via G-protein-coupled estrogen receptor 1 (GPER1) in rat astroglial cell line (C6). The C6 cell death induced by Aß was decreased by OBHS (1 µM) treatment for 45 min. This rapid protective action was blocked by GPER1 specific antagonist or siRNA knockdown. Inhibitors of phosphatidylinositol 3-kinase (PI3k)/Akt and extracellular signal-regulated kinase (ERK) activation also exhibited similar effects as GPER1 antagonist in blocking the protective effects of OBHS. Moreover, the expression of anti-apoptotic protein Bcl-2 was also increased by OBHS as a consequence of the activation of GPER1-PI3K/Akt and ERK pathways. Additionally, the phenyl sulfonate moiety of OBHS played a vital role in producing GPER1's agonist property according to the molecular docking analysis. These findings suggest that OBHS provide protection directed at enhancing glial cell survival through the activation of GPER1, which, in turn, offers a novel insight into the molecular mechanisms behind the potential application of OBHS in treating Alzheimer's disease (AD).


Subject(s)
Amyloid beta-Peptides/toxicity , Estrogen Receptor Antagonists/pharmacology , MAP Kinase Signaling System/physiology , Peptide Fragments/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Estrogen Receptor Antagonists/chemistry , MAP Kinase Signaling System/drug effects , Protein Structure, Secondary , Rats , Receptors, G-Protein-Coupled/agonists
16.
Bioorg Med Chem ; 25(13): 3531-3539, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28506582

ABSTRACT

A strategy by integrating biological imaging into early stages of the drug discovery process can improve our understanding of drug activity during preclinical and clinical study. In this article, we designed and synthesized coumarin-based nonsteroidal type fluorescence ligands for drug-target binding imaging. Among these synthesized compounds, 3e, 3f and 3h showed potent ER binding affinity and 3e (IC50=0.012µM) exhibited excellent ERα antagonistic activity, its antiproliferative potency in breast cancer MCF-7 cells is equipotent to the approved drug tamoxifen. The fluorescence of compounds 3e and 3f depended on the solvent properties and showed significant changes when mixed with ERα or ERß in vitro. Furthermore, target molecule 3e could cross the cell membrane, localize and image drug-target interaction in real time without cell washing. Thus, the coumarin-based platform represents a promising new ER-targeted delivery vehicle with potential imaging and therapeutic properties.


Subject(s)
Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Coumarins/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Fluorescent Dyes/pharmacology , Small Molecule Libraries/pharmacology , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Coumarins/chemical synthesis , Coumarins/chemistry , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , Female , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , MCF-7 Cells , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Tamoxifen/chemistry , Tamoxifen/pharmacology
17.
Chem Rec ; 16(2): 897-906, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26969213

ABSTRACT

This review seeks to provide coverage on the recent advances in chiral squaramide-catalyzed asymmetric transformations and their applications in the synthesis of a variety of chiral biologically active compounds. It aims to give an overview highlighting the new reaction types and enantioenriched medicinal scaffolds developed in the last few years.


Subject(s)
Quinine/analogs & derivatives , Catalysis , Humans , Quinine/chemistry , Stereoisomerism
18.
Bioorg Med Chem ; 24(13): 3062-3074, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27240467

ABSTRACT

Breast cancer is the most frequent cancer in women worldwide, and incidence is increasing year by year. Although current selective estrogen receptor modulators (SERMs) have clear advantages in the treatment of hormone-responsive breast cancer, they are ineffective for ER(-). In this study, we describe the design and synthesis of a series of dual-acting estrogen receptor (ER) and histone deacetylase (HDAC) inhibitors with incorporation of the ferrocenyl moiety, leading to novel hybrid ferrocenyl complexes (FcOBHS-HDACis) for breast cancer therapy. It is worth to note that these ferrocenyl conjugates could not only potently inhibit HDACs and the proliferation of ERα positive (ER(+)) breast cancer cells (MCF-7), but also show significant antiproliferative effect on ER(-) breast cancer cells (MDA-MB-231). Thus, the FcOBHS-HDACi conjugates represent a novel approach to the development of efficiently dual-acting agents for treatment of breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Bridged Bicyclo Compounds/chemistry , Bridged Bicyclo Compounds/pharmacology , Ferrous Compounds/chemistry , Ferrous Compounds/pharmacology , Binding Sites , Breast Neoplasms/physiopathology , Bridged Bicyclo Compounds/chemical synthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Female , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Inhibitory Concentration 50 , Models, Biological , Molecular Structure , Protein Binding/drug effects , Receptors, Estrogen/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL