Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 268(Pt 2): 131898, 2024 May.
Article in English | MEDLINE | ID: mdl-38677680

ABSTRACT

Diabetic nephropathy (DN) is one of the most severe complications of diabetes mellitus. Succinate Receptor 1 (SUCNR1), a member of the G-protein-coupled receptor (GPCR) family, represents a potential target for treatment of DN. Here, utilizing multi-strategy in silico virtual screening methods containing AlphaFold2 modelling, molecular dynamics (MD) simulation, ligand-based pharmacophore screening, molecular docking and machine learning-based similarity clustering, we successfully identified a novel antagonist of SUCNR1, AK-968/12117473 (Cpd3). Through extensive in vitro experiments, including dual-luciferase reporter assay, cellular thermal shift assay, immunofluorescence, and western blotting, we substantiated that Cpd3 could specifically target SUCNR1, inhibit the activation of NF-κB pathway, and ameliorate epithelial-mesenchymal transition (EMT) and extracellular matrix (ECM) deposition in renal tubular epithelial cells (NRK-52E) under high glucose conditions. Further in silico simulations revealed the molecular basis of the SUCNR1-Cpd3 interaction, and the in vitro metabolic stability assay indicated favorable drug-like pharmacokinetic properties of Cpd3. This work not only successfully pinpointed Cpd3 as a specific antagonist of SUCNR1 to serve as a promising candidate in the realm of therapeutic interventions for DN, but also provides a paradigm of dry-wet combined discovery strategies for GPCR-based therapeutics.


Subject(s)
Diabetic Nephropathies , Molecular Docking Simulation , Molecular Dynamics Simulation , Receptors, G-Protein-Coupled , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Humans , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Epithelial-Mesenchymal Transition/drug effects , Computer Simulation , Drug Discovery , NF-kappa B/metabolism , NF-kappa B/antagonists & inhibitors , Cell Line , Animals , Signal Transduction/drug effects
2.
Nat Commun ; 15(1): 1009, 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38307859

ABSTRACT

Tumor-secreted factors contribute to the development of a microenvironment that facilitates the escape of cancer cells from immunotherapy. In this study, we conduct a retrospective comparison of the proteins secreted by hepatocellular carcinoma (HCC) cells in responders and non-responders among a cohort of ten patients who received Nivolumab (anti-PD-1 antibody). Our findings indicate that non-responders have a high abundance of secreted RNase1, which is associated with a poor prognosis in various cancer types. Furthermore, mice implanted with HCC cells that overexpress RNase1 exhibit immunosuppressive tumor microenvironments and diminished response to anti-PD-1 therapy. RNase1 induces the polarization of macrophages towards a tumor growth-promoting phenotype through activation of the anaplastic lymphoma kinase (ALK) signaling pathway. Targeting the RNase1/ALK axis reprograms the macrophage polarization, with increased CD8+ T- and Th1- cell recruitment. Moreover, simultaneous targeting of the checkpoint protein PD-1 unleashes cytotoxic CD8+ T-cell responses. Treatment utilizing both an ALK inhibitor and an anti-PD-1 antibody exhibits enhanced tumor regression and facilitates long-term immunity. Our study elucidates the role of RNase1 in mediating tumor resistance to immunotherapy and reveals an RNase1-mediated immunosuppressive tumor microenvironment, highlighting the potential of targeting RNase1 as a promising strategy for cancer immunotherapy in HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Humans , Mice , Anaplastic Lymphoma Kinase , Carcinoma, Hepatocellular/metabolism , CD8-Positive T-Lymphocytes , Immunosuppression Therapy , Liver Neoplasms/metabolism , Retrospective Studies , Ribonucleases , Tumor Microenvironment
3.
Angew Chem Int Ed Engl ; 62(48): e202313429, 2023 11 27.
Article in English | MEDLINE | ID: mdl-37840440

ABSTRACT

The oxidosqualene cyclase (OSC) catalyzed cyclization of the linear substrate (3S)-2,3-oxidosqualene to form diverse pentacyclic triterpenoid (PT) skeletons is one of the most complex reactions in nature. Friedelin has a unique PT skeleton involving a fascinating nine-step cation shuttle run (CSR) cascade rearrangement reaction, in which the carbocation formed at C2 moves to the other side of the skeleton, runs back to C3 to yield a friedelin cation, which is finally deprotonated. However, as crystal structure data of plant OSCs are lacking, it remains unknown why the CSR cascade reactions occur in friedelin biosynthesis, as does the exact catalytic mechanism of the CSR. In this study, we determined the first cryogenic electron microscopy structure of a plant OSC, friedelin synthase, from Tripterygium wilfordii Hook. f (TwOSC). We also performed quantum mechanics/molecular mechanics simulations to reveal the energy profile for the CSR cascade reaction and identify key residues crucial for PT skeleton formation. Furthermore, we semirationally designed two TwOSC mutants, which significantly improved the yields of friedelin and ß-amyrin, respectively.


Subject(s)
Intramolecular Transferases , Triterpenes , Triterpenes/chemistry , Intramolecular Transferases/genetics , Catalysis , Cations
5.
Oncogene ; 40(31): 4992-5001, 2021 08.
Article in English | MEDLINE | ID: mdl-34172932

ABSTRACT

Programmed cell death 1 (PD-1) is widely expressed in tumor-infiltrating lymphocytes (TILs) of triple-negative breast cancer (TNBC). As a dominant inhibitory immune checkpoint (ICP) receptor, cell surface PD-1 is well-known to transduce negative signaling of effector T cell activity during cell-cell contact. However, despite its well-documented inhibitory effects, higher PD-1 expression in TILs is significantly associated with longer survival in TNBC patients. This phenomenon raises an interesting question whether PD-1 harbors positive activity to enhance anti-tumor immunity. Here, we show that PD-1 is secreted in an exosomal form by activated T cells and can remotely interact with either cell surface or exosomal programmed death-ligand 1 (PD-L1), induce PD-L1 internalization via clathrin-mediated endocytosis, and thereby prevent subsequent cellular PD-L1: PD-1 interaction, restoring tumor surveillance through attenuating PD-L1-induced suppression of tumor-specific cytotoxic T cell activity. Our results, through revealing an anti-PD-L1 function of exosomal PD-1, provide a positive role to enhance cytotoxic T cell activity and a potential therapeutic strategy of modifying the exosome surface with membrane-bound inhibitory ICP receptors to attenuate the suppressive tumor immune microenvironment.


Subject(s)
B7-H1 Antigen/metabolism , Exosomes/metabolism , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Triple Negative Breast Neoplasms/etiology , Triple Negative Breast Neoplasms/metabolism , Animals , B7-H1 Antigen/genetics , Biomarkers , Cytotoxicity, Immunologic , Disease Susceptibility , Exosomes/ultrastructure , Female , Humans , Immunomodulation , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Models, Biological , Programmed Cell Death 1 Receptor/genetics , T-Lymphocyte Subsets/pathology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
6.
Front Chem ; 9: 689731, 2021.
Article in English | MEDLINE | ID: mdl-34150721

ABSTRACT

The catalytic promiscuity and fidelity of cytochrome P450 enzymes are widespread in the skeletal modification of terpenoid natural products and have attracted much attention. CYP76AH1 is involved in key modification reactions in the biosynthetic pathway of tanshinone, a well-known medicinal norditerpenoid. In this work, classical molecular dynamic simulations, metadynamics, and DFT calculations were performed to investigate the protein conformational dynamics, ligand binding poses, and catalytic reaction mechanism in wide-type and mutant CYP76AH1. Our results not only reveal a plausible enzymatic mechanism for mutant CYP76AH1 leading to various products but also provide valuable guidance for rational protein engineering of the CYP76 family.

7.
Nat Commun ; 12(1): 2788, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33986289

ABSTRACT

Human ribonuclease 1 (hRNase 1) is critical to extracellular RNA clearance and innate immunity to achieve homeostasis and host defense; however, whether it plays a role in cancer remains elusive. Here, we demonstrate that hRNase 1, independently of its ribonucleolytic activity, enriches the stem-like cell population and enhances the tumor-initiating ability of breast cancer cells. Specifically, secretory hRNase 1 binds to and activates the tyrosine kinase receptor ephrin A4 (EphA4) signaling to promote breast tumor initiation in an autocrine/paracrine manner, which is distinct from the classical EphA4-ephrin juxtacrine signaling through contact-dependent cell-cell communication. In addition, analysis of human breast tumor tissue microarrays reveals a positive correlation between hRNase 1, EphA4 activation, and stem cell marker CD133. Notably, high hRNase 1 level in plasma samples is positively associated with EphA4 activation in tumor tissues from breast cancer patients, highlighting the pathological relevance of the hRNase 1-EphA4 axis in breast cancer. The discovery of hRNase 1 as a secretory ligand of EphA4 that enhances breast cancer stemness suggests a potential treatment strategy by inactivating the hRNase 1-EphA4 axis.


Subject(s)
Breast Neoplasms/pathology , Carcinogenesis/pathology , Ephrin-A4/metabolism , Neoplastic Stem Cells/pathology , Ribonuclease, Pancreatic/metabolism , AC133 Antigen/metabolism , Animals , Breast Neoplasms/genetics , Carcinogenesis/genetics , Cell Line , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Protein Binding/genetics , Ribonuclease, Pancreatic/blood , Ribonuclease, Pancreatic/genetics , Treatment Outcome
8.
Front Oncol ; 11: 615427, 2021.
Article in English | MEDLINE | ID: mdl-33833984

ABSTRACT

The expression of Centromere Protein U (CENP-U) is closely related to tumor malignancy. Till now, the role of CENP-U in the malignant progression of breast cancer remains unclear. In this study, we found that CENP-U protein was highly expressed in the primary invasive breast cancer tissues compared to the paired adjacent histologically normal tissues and ductal carcinoma in situ (DCIS) tissues. After CENP-U was knocked down, the proliferation and colony-forming abilities of breast cancer cells were significantly suppressed, whereas the portion of apoptotic cells was increased. Meanwhile, the PI3K/AKT/NF-κB pathway was significantly inhibited. In vivo studies showed that, the inhibition of CENP-U repressed the tumor growth in orthotopic breast cancer models. Therefore, our study demonstrated that the CENP-U might act as an oncogene and promote breast cancer progression via activation of the PI3K/AKT/NF-κB pathway, which suggests a promising direction for targeting therapy in breast cancer.

9.
J Hepatol ; 74(4): 907-918, 2021 04.
Article in English | MEDLINE | ID: mdl-33031845

ABSTRACT

BACKGROUND & AIMS: There are currently limited therapeutic options for hepatocellular carcinoma (HCC), particularly when it is diagnosed at advanced stages. Herein, we examined the pathophysiological role of ROS1 and assessed the utility of ROS1-targeted therapy for the treatment of HCC. METHODS: Recombinant ribonucleases (RNases) were purified, and the ligand-receptor relationship between RNase7 and ROS1 was validated in HCC cell lines by Duolink, immunofluorescence, and immunoprecipitation assays. Potential interacting residues between ROS1 and RNase7 were predicted using a protein-protein docking approach. The oncogenic function of RNase7 was analyzed by cell proliferation, migration and invasion assays, and a xenograft mouse model. The efficacy of anti-ROS1 inhibitor treatment was evaluated in patient-derived xenograft (PDX) and orthotopic models. Two independent patient cohorts were analyzed to evaluate the pathological relevance of RNase7/ROS1. RESULTS: RNase7 associated with ROS1's N3-P2 domain and promoted ROS1-mediated oncogenic transformation. Patients with HCC exhibited elevated plasma RNase7 levels compared with healthy individuals. High ROS1 and RNase7 expression were strongly associated with poor prognosis in patients with HCC. In both HCC PDX and orthotopic mouse models, ROS1 inhibitor treatment markedly suppressed RNase7-induced tumorigenesis, leading to decreased plasma RNase7 levels and tumor shrinkage in mice. CONCLUSIONS: RNase7 serves as a high-affinity ligand for ROS1. Plasma RNase7 could be used as a biomarker to identify patients with HCC who may benefit from anti-ROS1 treatment. LAY SUMMARY: Receptor tyrosine kinases are known to be involved in tumorigenesis and have been targeted therapeutically for a number of cancers, including hepatocellular carcinoma. ROS1 is the only such receptor with kinase activity whose ligand has not been identified. Herein, we show that RNase7 acts as a ligand to activate ROS1 signaling. This has important pathophysiological and therapeutic implications. Anti-ROS1 inhibitors could be used to treatment patients with hepatocellular carcinoma and high RNase7 levels.


Subject(s)
Carcinogenesis , Carcinoma, Hepatocellular , Crizotinib/pharmacology , Liver Neoplasms , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Ribonucleases/metabolism , Animals , Biomarkers, Tumor/metabolism , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Migration Assays/methods , Cell Proliferation/drug effects , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Mice , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
10.
J Chem Inf Model ; 60(4): 2082-2090, 2020 04 27.
Article in English | MEDLINE | ID: mdl-32286817

ABSTRACT

Natural products are the major resource of drug discovery, and terpenoids represent the largest family of natural products. Terpenome is defined as all terpenoid-like and terpenoid-derived natural compounds, including the terpenoids, steroids, and their derivatives. Herein, aiming to navigate the chemical and biological space of terpenome, the first comprehensive database dedicated to terpenome research has been developed by collecting over 110 000 terpenome molecules from various resources, distributed in 14 351 species, belonging to 1109 families, and showing activity against 1366 biological targets. Much of the publically available information or computationally predicted properties for each terpenome molecule is annotated and integrated into TeroKit (http://terokit.qmclab.com/), serving as free Web server for academic use. Moreover, several practical toolkits, such as target profiling and conformer generation modules, are also implemented to facilitate the drug discovery of terpenome.


Subject(s)
Biological Products , Terpenes , Computers , Databases, Factual , Drug Discovery
11.
Cancer Res ; 80(11): 2298-2310, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32156778

ABSTRACT

Immunotherapies targeting programmed cell death protein 1 (PD-1) and programmed cell death 1 ligand 1 (PD-L1) immune checkpoints represent a major breakthrough in cancer treatment. PD-1 is an inhibitory receptor expressed on the surface of activated T cells that dampens T-cell receptor (TCR)/CD28 signaling by engaging with its ligand PD-L1 expressed on cancer cells. Despite the clinical success of PD-1 blockade using mAbs, most patients do not respond to the treatment, and the underlying regulatory mechanisms of PD-1 remain incompletely defined. Here we show that PD-1 is extensively N-glycosylated in T cells and the intensities of its specific glycoforms are altered upon TCR activation. Glycosylation was critical for maintaining PD-1 protein stability and cell surface localization. Glycosylation of PD-1, especially at the N58 site, was essential for mediating its interaction with PD-L1. The mAb STM418 specifically targeted glycosylated PD-1, exhibiting higher binding affinity to PD-1 than FDA-approved PD-1 antibodies, potently inhibiting PD-L1/PD-1 binding, and enhancing antitumor immunity. Together, these findings provide novel insights into the functional significance of PD-1 glycosylation and offer a rationale for targeting glycosylated PD-1 as a potential strategy for immunotherapy. SIGNIFICANCE: These findings demonstrate that glycosylation of PD-1 is functionally significant and targeting glycosylated PD-1 may serve as a means to improve immunotherapy response.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Leukemia, T-Cell/drug therapy , Leukemia, T-Cell/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/metabolism , Female , Glycosylation , HEK293 Cells , Heterografts , Humans , Jurkat Cells , Leukemia, T-Cell/metabolism , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy , Nivolumab/pharmacology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
12.
Cancer Lett ; 482: 102-111, 2020 07 10.
Article in English | MEDLINE | ID: mdl-31705927

ABSTRACT

Triple-negative breast cancer (TNBC) is characterized by high vascularity, but anti-angiogenic therapies show poor efficacy. Centromere protein U (CENPU), a centromere component essential for mitosis, is associated with tumorigenesis in multiple cancers; however, little is known of its role in breast cancer. Here, we investigate its expression and function of promoting angiogenesis in TNBC. Immunohistochemical staining revealed high CENPU expression in TNBC tissue and high CENPU levels correlated significantly with poor distant metastasis-free and overall survival. Knockdown of CENPU in TNBC cells inhibited vascular endothelial growth factor A (VEGFA) production and significantly reduced tube formation and migration of human umbilical vein endothelial cells in vitro. In a mouse xenograft model, CENPU knockdown reduced TNBC tumor growth concomitant with a reduction in CD31 + microvessel density. Mechanistic studies revealed that CENPU promoted angiogenesis by inhibiting the ubiquitination and proteasomal degradation of cyclooxygenase-2 (COX-2), leading to increased activation of the COX-2-p-ERK-HIF-1α-VEGFA signaling pathway. Taken together, our results demonstrate a critical role for CENPU in COX-2-mediated signaling for angiogenesis, and identify a role of CENPU in regulating angiogenesis in TNBC.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cyclooxygenase 2/metabolism , Histones/genetics , Histones/metabolism , Triple Negative Breast Neoplasms/pathology , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/cytology , Humans , MCF-7 Cells , Mice , Neoplasm Metastasis , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Ubiquitin/metabolism
14.
Cancer Res ; 79(4): 819-829, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30573522

ABSTRACT

PARP1 inhibitors (PARPi) are currently used in the clinic for the treatment of ovarian and breast cancers, yet their therapeutic efficacy against hepatocellular carcinoma (HCC) has been disappointing. To ensure therapeutic efficacy of PARPi against HCC, a disease often diagnosed at intermediate to advanced stages with no effective treatment options, it is critical to identify not only biomarkers to predict PARPi resistance but also rational treatments to overcome this. Here, we report that a heterodimer of EGFR and MET interacts with and phosphorylates Y907 of PARP1 in the nucleus, which contributes to PARPi resistance. Inhibition of both EGFR and MET sensitized HCC cells to PARPi, and both EGFR and MET are known to be overexpressed in HCC. This report provides an explanation for the poor efficacy of PARPi against HCC and suggests combinatorial treatment consisting of EGFR, MET, and PARP inhibitors may be an effective therapeutic strategy in HCC. SIGNIFICANCE: Regulation of PARP by the c-MET and EGFR heterodimer suggests a potentially effective combination therapy to sensitize HCC to PARPi.


Subject(s)
Carcinoma, Hepatocellular/pathology , Drug Resistance, Neoplasm , Liver Neoplasms/pathology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Proto-Oncogene Proteins c-met/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Am J Cancer Res ; 8(9): 1837-1846, 2018.
Article in English | MEDLINE | ID: mdl-30323975

ABSTRACT

Triple-negative breast cancer (TNBC), the most difficult-to-treat breast cancer subtype, lacks well-defined molecular targets. TNBC has increased programmed death-ligand 1 (PD-L1) expression, and its immunosuppressive nature makes it suitable for immune checkpoint blockade therapy. However, the response rate of TNBC to anti-PD-L1 or anti-programmed cell death protein 1 (PD-1) therapy remains unsatisfactory, as only 10-20% of TNBC patients have a partial response. Glycosylated PD-L1, the functional form of PD-L1, is required for PD-L1-PD-1 interaction. TNBC cells have significantly higher levels of glycosylated PD-L1 than non-TNBC cells do. In a screening of glucose analogs to block PD-L1 glycosylation, we found that 2-deoxyglucose (2-DG) can act as a glucose analog to decrease PD-L1 glycosylation. Because PARP inhibition upregulates PD-L1, 2-DG reduced PARP inhibition-mediated expression of glycosylated PD-L1. The combination of PARP inhibition and 2-DG had potent anti-tumor activity. Together, our results provide a strong rationale for investigating the targeting of PD-L1 glycosylation in TNBC further.

16.
Cancer Sci ; 108(6): 1177-1184, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28301080

ABSTRACT

Hormone therapy has become one of the main strategies for breast cancer, however, many estrogen receptor (ER) positive patients end in tumor collapse due to initial or acquired resistance to hormone treatment, which includes Fulvestrant. Here we report that ErbB receptors and downstream PI3K/AKT and ERK pathway have been reactivated after treatment of Fulvestrant in ER positive MCF-7 and T47D cells, which are related to Fulvestrant resistance. HSP90 is a universally expressed chaperone protein and plays a vital role in both normal and cancer cells, HSP90 inhibitor AUY922 can reverse this feedback reactivation effect of Fulvestrant by targeting multiple proteins related in ErbB receptors, PI3K/AKT and ERK pathway, which is much better than single targeting inhibitors. We also consolidate these effects in human fresh breast tumors. Combination of AUY922 and Fulvestrant may become a promising therapy strategy in breast cancer treatment.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Estradiol/analogs & derivatives , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Resorcinols/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Estradiol/pharmacology , Female , Fulvestrant , Humans , MCF-7 Cells , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Estrogen/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...