Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Clin Exp Med ; 23(8): 5445-5461, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37935952

ABSTRACT

The PI3K pathway is one of the most frequently altered signaling pathways in human cancer. In addition to its function in cancer cells, PI3K plays a complex role in modulating anti-tumor immune responses upon immune checkpoint inhibition (ICI). Here, we evaluated the effects of the pan-Class I PI3K inhibitor copanlisib on different immune cell types in vitro and on tumor growth and immune cell infiltration in syngeneic murine cancer models. Intermittent treatment with copanlisib resulted in a strong in vivo anti-tumor efficacy, increased tumor infiltration of activated T cells and macrophages, and increased CD8+ T cell/regulatory T cell and M1/M2 macrophage ratios. The strong in vivo efficacy was at least partially due to immunomodulatory activity of copanlisib, as in vitro these murine cancer cells were resistant to PI3K inhibition. Furthermore, the combination of copanlisib with the ICI antibody anti-PD-1 demonstrated enhanced anti-tumor efficacy in both ICI-sensitive and insensitive syngeneic mouse tumor models. Importantly, in an ICI-sensitive model, combination therapy resulted in complete remission and prevention of tumor recurrence. Thus, the combination of ICIs with PI3K inhibition by intermittently dosed copanlisib represents a promising new strategy to increase sensitivity to ICI therapies and to treat human solid cancers.


Subject(s)
Neoplasms , Phosphatidylinositol 3-Kinases , Humans , Animals , Mice , Phosphatidylinositol 3-Kinases/metabolism , T-Lymphocytes, Regulatory/metabolism , Neoplasms/drug therapy , Immunity , Tumor Microenvironment
2.
Cancer Cell ; 40(9): 939-956.e16, 2022 09 12.
Article in English | MEDLINE | ID: mdl-35985343

ABSTRACT

Mutations affecting isocitrate dehydrogenase (IDH) enzymes are prevalent in glioma, leukemia, and other cancers. Although mutant IDH inhibitors are effective against leukemia, they seem to be less active in aggressive glioma, underscoring the need for alternative treatment strategies. Through a chemical synthetic lethality screen, we discovered that IDH1-mutant glioma cells are hypersensitive to drugs targeting enzymes in the de novo pyrimidine nucleotide synthesis pathway, including dihydroorotate dehydrogenase (DHODH). We developed a genetically engineered mouse model of mutant IDH1-driven astrocytoma and used it and multiple patient-derived models to show that the brain-penetrant DHODH inhibitor BAY 2402234 displays monotherapy efficacy against IDH-mutant gliomas. Mechanistically, this reflects an obligate dependence of glioma cells on the de novo pyrimidine synthesis pathway and mutant IDH's ability to sensitize to DNA damage upon nucleotide pool imbalance. Our work outlines a tumor-selective, biomarker-guided therapeutic strategy that is poised for clinical translation.


Subject(s)
Brain Neoplasms , Glioma , Leukemia , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Enzyme Inhibitors/therapeutic use , Glioma/drug therapy , Glioma/genetics , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mice , Mutation , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Salicylanilides , Triazoles
3.
Leukemia ; 33(10): 2403-2415, 2019 10.
Article in English | MEDLINE | ID: mdl-30940908

ABSTRACT

Acute myeloid leukemia (AML) is a devastating disease, with the majority of patients dying within a year of diagnosis. For patients with relapsed/refractory AML, the prognosis is particularly poor with currently available treatments. Although genetically heterogeneous, AML subtypes share a common differentiation arrest at hematopoietic progenitor stages. Overcoming this differentiation arrest has the potential to improve the long-term survival of patients, as is the case in acute promyelocytic leukemia (APL), which is characterized by a chromosomal translocation involving the retinoic acid receptor alpha gene. Treatment of APL with all-trans retinoic acid (ATRA) induces terminal differentiation and apoptosis of leukemic promyelocytes, resulting in cure rates of over 80%. Unfortunately, similarly efficacious differentiation therapies have, to date, been lacking outside of APL. Inhibition of dihydroorotate dehydrogenase (DHODH), a key enzyme in the de novo pyrimidine synthesis pathway, was recently reported to induce differentiation of diverse AML subtypes. In this report we describe the discovery and characterization of BAY 2402234 - a novel, potent, selective and orally bioavailable DHODH inhibitor that shows monotherapy efficacy and differentiation induction across multiple AML subtypes. Herein, we present the preclinical data that led to initiation of a phase I evaluation of this inhibitor in myeloid malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Enzyme Inhibitors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Dihydroorotate Dehydrogenase , Female , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Pyrimidines/metabolism , THP-1 Cells , Translocation, Genetic/drug effects
4.
Nat Commun ; 9(1): 2068, 2018 05 25.
Article in English | MEDLINE | ID: mdl-29802342

ABSTRACT

Transient activation of Src oncoprotein in non-transformed, breast epithelial cells can initiate an epigenetic switch to the stably transformed state via a positive feedback loop that involves the inflammatory transcription factors STAT3 and NF-κB. Here, we develop an experimental and computational pipeline that includes 1) a Bayesian network model (AccessTF) that accurately predicts protein-bound DNA sequence motifs based on chromatin accessibility, and 2) a scoring system (TFScore) that rank-orders transcription factors as candidates for being important for a biological process. Genetic experiments validate TFScore and suggest that more than 40 transcription factors contribute to the oncogenic state in this model. Interestingly, individual depletion of several of these factors results in similar transcriptional profiles, indicating that a complex and interconnected transcriptional network promotes a stable oncogenic state. The combined experimental and computational pipeline represents a general approach to comprehensively identify transcriptional regulators important for a biological process.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/genetics , Transcription Factors/metabolism , Breast/cytology , Breast/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Regulatory Networks/genetics , Genome, Human/genetics , Humans , Models, Genetic , Transcription Factors/genetics
5.
Exp Cell Res ; 348(2): 123-131, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27646113

ABSTRACT

Cell migration is a central process in the development and maintenance of multicellular organisms. Tissue formation during embryonic development, wound healing, immune responses and invasive tumors all require the orchestrated movement of cells to specific locations. Histone demethylase proteins alter transcription by regulating the chromatin state at specific gene loci. FBXL10 is a conserved and ubiquitously expressed member of the JmjC domain-containing histone demethylase family and is implicated in the demethylation of H3K4me3 and H3K36me2 and thereby removing active chromatin marks. However, the physiological role of FBXL10 in vivo remains largely unknown. Therefore, we established an inducible gain of function model to analyze the role of Fbxl10 and compared wild-type with Fbxl10 overexpressing mouse embryonic fibroblasts (MEFs). Our study shows that overexpression of Fbxl10 in MEFs doesn't influence the proliferation capability but leads to an enhanced migration capacity in comparison to wild-type MEFs. Transcriptome and ChIP-seq experiments demonstrated that Fbxl10 binds to genes involved in migration like Areg, Mdk, Lmnb1, Thbs1, Mgp and Cxcl12. Taken together, our results strongly suggest that Fbxl10 plays a critical role in migration by binding to the promoter region of migration-associated genes and thereby might influences cell behaviour to a possibly more aggressive phenotype.


Subject(s)
Cell Movement , Embryo, Mammalian/cytology , F-Box Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Animals , Cell Movement/drug effects , Cell Movement/genetics , Doxycycline/pharmacology , F-Box Proteins/genetics , Female , Fibroblasts/drug effects , Gene Expression Profiling , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Mice, Inbred C57BL , Mice, Transgenic , RNA, Small Interfering/metabolism , Transcription, Genetic/drug effects , Transcriptome/drug effects , Transcriptome/genetics , Transgenes
6.
Cell ; 167(1): 171-186.e15, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27641501

ABSTRACT

While acute myeloid leukemia (AML) comprises many disparate genetic subtypes, one shared hallmark is the arrest of leukemic myeloblasts at an immature and self-renewing stage of development. Therapies that overcome differentiation arrest represent a powerful treatment strategy. We leveraged the observation that the majority of AML, despite their genetically heterogeneity, share in the expression of HoxA9, a gene normally downregulated during myeloid differentiation. Using a conditional HoxA9 model system, we performed a high-throughput phenotypic screen and defined compounds that overcame differentiation blockade. Target identification led to the unanticipated discovery that inhibition of the enzyme dihydroorotate dehydrogenase (DHODH) enables myeloid differentiation in human and mouse AML models. In vivo, DHODH inhibitors reduced leukemic cell burden, decreased levels of leukemia-initiating cells, and improved survival. These data demonstrate the role of DHODH as a metabolic regulator of differentiation and point to its inhibition as a strategy for overcoming differentiation blockade in AML.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Molecular Targeted Therapy , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Cell Differentiation , Dihydroorotate Dehydrogenase , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , High-Throughput Screening Assays , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Myeloid Cells/pathology , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Pyrimidines/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/isolation & purification , Small Molecule Libraries/therapeutic use , Xenograft Model Antitumor Assays
7.
Proc Natl Acad Sci U S A ; 112(18): 5708-13, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25902495

ABSTRACT

Colony formation in soft agar is the gold-standard assay for cellular transformation in vitro, but it is unsuited for high-throughput screening. Here, we describe an assay for cellular transformation that involves growth in low attachment (GILA) conditions and is strongly correlated with the soft-agar assay. Using GILA, we describe high-throughput screens for drugs and genes that selectively inhibit or increase transformation, but not proliferation. Such molecules are unlikely to be found through conventional drug screening, and they include kinase inhibitors and drugs for noncancer diseases. In addition to known oncogenes, the genetic screen identifies genes that contribute to cellular transformation. Lastly, we demonstrate the ability of Food and Drug Administration-approved noncancer drugs to selectively kill ovarian cancer cells derived from patients with chemotherapy-resistant disease, suggesting this approach may provide useful information for personalized cancer treatment.


Subject(s)
Agar/chemistry , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Ovarian Neoplasms/drug therapy , Adenosine Triphosphate/chemistry , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic , Chemistry, Pharmaceutical , Drug Design , Female , Fibroblasts/pathology , Flow Cytometry , High-Throughput Screening Assays , Humans , Open Reading Frames , Ovarian Neoplasms/pathology
8.
Proc Natl Acad Sci U S A ; 111(29): 10574-9, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25002509

ABSTRACT

Metformin, a first-line diabetes drug linked to cancer prevention in retrospective clinical analyses, inhibits cellular transformation and selectively kills breast cancer stem cells (CSCs). Although a few metabolic effects of metformin and the related biguanide phenformin have been investigated in established cancer cell lines, the global metabolic impact of biguanides during the process of neoplastic transformation and in CSCs is unknown. Here, we use LC/MS/MS metabolomics (>200 metabolites) to assess metabolic changes induced by metformin and phenformin in an Src-inducible model of cellular transformation and in mammosphere-derived breast CSCs. Although phenformin is the more potent biguanide in both systems, the metabolic profiles of these drugs are remarkably similar, although not identical. During the process of cellular transformation, biguanide treatment prevents the boost in glycolytic intermediates at a specific stage of the pathway and coordinately decreases tricarboxylic acid (TCA) cycle intermediates. In contrast, in breast CSCs, biguanides have a modest effect on glycolytic and TCA cycle intermediates, but they strongly deplete nucleotide triphosphates and may impede nucleotide synthesis. These metabolic profiles are consistent with the idea that biguanides inhibit mitochondrial complex 1, but they indicate that their metabolic effects differ depending on the stage of cellular transformation.


Subject(s)
Citric Acid Cycle/drug effects , Glycolysis/drug effects , Metformin/pharmacology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nucleotides/metabolism , Phenformin/pharmacology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/metabolism , Biguanides/pharmacology , Cell Line, Transformed , Cell Line, Tumor , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Folic Acid/metabolism , Glycerophosphates/metabolism , Humans , Lactates/metabolism , Metabolome/drug effects , Neoplastic Stem Cells/drug effects , Ribonucleotides/metabolism , Tamoxifen/pharmacology , src-Family Kinases/metabolism
9.
J Biol Chem ; 287(37): 30984-92, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22825849

ABSTRACT

Fbxl10 (Jhdm1b/Kdm2b) is a conserved and ubiquitously expressed member of the JHDM (JmjC domain-containing histone demethylase) family. Fbxl10 was implicated in the demethylation of H3K4me3 or H3K36me2 thereby removing active chromatin marks and inhibiting gene transcription. Apart from the JmjC domain, Fbxl10 consists of a CxxC domain, a PHD domain, and an Fbox domain. By purifying the JmjC and the PHD domain of Fbxl10 and using different approaches we were able to characterize the properties of these domains in vitro. Our results suggest that Fbxl10 is rather a H3K4me3 than a H3K36me2 histone demethylase. The PHD domain exerts a dual function in binding H3K4me3 and H3K36me2 and exhibiting E3 ubiquitin ligase activity. We generated mouse embryonic fibroblasts stably overexpressing Fbxl10. These cells reveal an increase in cell size but no changes in proliferation, mitosis, or apoptosis. Using a microarray approach we were able to identify potentially new target genes for Fbxl10 including chemokines, the noncoding RNA Xist, and proteins involved in metabolic processes. Additionally, we found that Fbxl10 is recruited to the promoters of Ccl7, Xist, Crabp2, and RipK3. Promoter occupancy by Fbxl10 was accompanied by reduced levels of H3K4me3 but unchanged levels of H3K36me2. Furthermore, knockdown of Fbxl10 using small interfering RNA approaches showed inverse regulation of Fbxl10 target genes. In summary, our data reveal a regulatory role of Fbxl10 in cell morphology, chemokine expression, and the metabolic control of fibroblasts.


Subject(s)
Chemokines/biosynthesis , F-Box Proteins/metabolism , Fibroblasts/metabolism , Gene Expression Regulation/physiology , Jumonji Domain-Containing Histone Demethylases/metabolism , Metabolome/physiology , Oxidoreductases, N-Demethylating/metabolism , Animals , Cells, Cultured , Chemokines/genetics , F-Box Proteins/genetics , Fibroblasts/cytology , Histones/genetics , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Oxidoreductases, N-Demethylating/genetics , Promoter Regions, Genetic/physiology , Protein Structure, Tertiary , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transcription, Genetic/physiology
10.
Biochem Biophys Res Commun ; 421(4): 665-70, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22542627

ABSTRACT

Histone modifying enzymes confer epigenetic marks, directing the changes in gene expression required for diverse cellular processes. Lysine-specific demethylase 1 (LSD1) functions as a transcriptional coregulator by demethylating histone H3 on lysine 4 and lysine 9. Analyzing transcriptomes on microarrays, we identified genes which represent inflammatory-related targets of LSD1. We demonstrate a repressive role of LSD1 in proinflammatory cytokine expression such as IL1α, IL1ß, IL6 and IL8 and classical complement components. Consistently, LSD1 occupies and regulates the promoter of these genes. In addition, we demonstrate that HDAC1 and LSD1 synergistically regulate these inflammatory-related genes. Our data reveal a novel role for LSD1 in suppressing immune responses.


Subject(s)
Complement Pathway, Classical/genetics , Cytokines/genetics , Epigenesis, Genetic , Histone Deacetylase 1/metabolism , Histone Demethylases/metabolism , Immunity/genetics , Inflammation/genetics , Cell Line, Tumor , Cytokines/antagonists & inhibitors , Down-Regulation , Gene Knockdown Techniques , Hep G2 Cells , Histone Deacetylase 1/genetics , Histone Demethylases/genetics , Humans
11.
Int J Cancer ; 131(11): 2704-9, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22447389

ABSTRACT

Post-translational modifications of histones by chromatin modifying enzymes regulate chromatin structure and gene expression. As deregulation of histone modifications contributes to cancer progression, inhibition of chromatin modifying enzymes such as histone demethylases is an attractive therapeutic strategy to impair cancer growth. Lysine-specific demethylase 1 (LSD1) removes mono- and dimethyl marks from lysine 4 or 9 of histone H3. LSD1 in association with the androgen receptor (AR) controls androgen-dependent gene expression and prostate tumor cell proliferation, thus highlighting LSD1 as a drug target. By combining protein structure similarity clustering and in vitro screening, we identified Namoline, a γ-pyrone, as a novel, selective and reversible LSD1 inhibitor. Namoline blocks LSD1 demethylase activity in vitro and in vivo. Inhibition of LSD1 by Namoline leads to silencing of AR-regulated gene expression and severely impairs androgen-dependent proliferation in vitro and in vivo. Thus, Namoline is a novel promising starting compound for the development of therapeutics to treat androgen-dependent prostate cancer.


Subject(s)
Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Pyrones/pharmacology , Androgens/metabolism , Animals , Cell Growth Processes/drug effects , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Male , Methylation/drug effects , Mice , Mice, Nude , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Processing, Post-Translational/drug effects , Receptors, Androgen/metabolism , Xenograft Model Antitumor Assays
12.
Biochem J ; 430(1): 87-95, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20518742

ABSTRACT

Matriptase-2 is a member of the TTSPs (type II transmembrane serine proteases), an emerging class of cell surface proteases involved in tissue homoeostasis and several human disorders. Matriptase-2 exhibits a domain organization similar to other TTSPs, with a cytoplasmic N-terminus, a transmembrane domain and an extracellular C-terminus containing the non-catalytic stem region and the protease domain. To gain further insight into the biochemical functions of matriptase-2, we characterized the subcellular localization of the monomeric and multimeric form and identified cell surface shedding as a defining point in its proteolytic processing. Using HEK (human embryonic kidney)-293 cells, stably transfected with cDNA encoding human matriptase-2, we demonstrate a cell membrane localization for the inactive single-chain zymogen. Membrane-associated matriptase-2 is highly N-glycosylated and occurs in monomeric, as well as multimeric, forms covalently linked by disulfide bonds. Furthermore, matriptase-2 undergoes shedding into the conditioned medium as an activated two-chain form containing the catalytic domain, which is cleaved at the canonical activation motif, but is linked to a released portion of the stem region via a conserved disulfide bond. Cleavage sites were identified by MS, sequencing and mutational analysis. Interestingly, cell surface shedding and activation of a matriptase-2 variant bearing a mutation at the active-site serine residue is dependent on the catalytic activity of co-expressed or co-incubated wild-type matriptase-2, indicating a transactivation and trans-shedding mechanism.


Subject(s)
Enzyme Precursors/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Catalysis , Catalytic Domain , Cell Line , Cell Membrane/metabolism , Culture Media, Conditioned , Enzyme Activation , Enzyme Precursors/genetics , Extracellular Space/enzymology , Humans , Membrane Proteins/genetics , Mutation , Protein Binding , Serine Endopeptidases/genetics , Transfection
13.
Carcinogenesis ; 31(3): 512-20, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20042638

ABSTRACT

Breast carcinogenesis is a multistep process involving both genetic and epigenetic changes. Since epigenetic changes like histone modifications are potentially reversible processes, much effort has been directed toward understanding this mechanism with the goal of finding novel therapies as well as more refined diagnostic and prognostic tools in breast cancer. Lysine-specific demethylase 1 (LSD1) plays a key role in the regulation of gene expression by removing the methyl groups from methylated lysine 4 of histone H3 and lysine 9 of histone H3. LSD1 is essential for mammalian development and involved in many biological processes. Considering recent evidence that LSD1 is involved in carcinogenesis, we investigated the role of LSD1 in breast cancer. Therefore, we developed an enzyme-linked immunosorbent assay to determine LSD1 protein levels in tissue specimens of breast cancer and measured very high LSD1 levels in estrogen receptor (ER)-negative tumors. Pharmacological LSD1 inhibition resulted in growth inhibition of breast cancer cells. Knockdown of LSD1 using small interfering RNA approach induced regulation of several proliferation-associated genes like p21, ERBB2 and CCNA2. Additionally, we found that LSD1 is recruited to the promoters of these genes. In summary, our data indicate that LSD1 may provide a predictive marker for aggressive biology and a novel attractive therapeutic target for treatment of ER-negative breast cancers.


Subject(s)
Adenocarcinoma/enzymology , Breast Neoplasms/enzymology , Histone Demethylases/physiology , Neoplasm Invasiveness/genetics , Neoplasm Proteins/physiology , Adenocarcinoma/chemistry , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Biomarkers, Tumor , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor/enzymology , Chromatin Immunoprecipitation , Cyclin A2/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Enzyme-Linked Immunosorbent Assay , Female , Genes, erbB-2 , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/biosynthesis , Histone Demethylases/genetics , Histones/analysis , Humans , Methylation , Neoplasm Proteins/analysis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Prognosis , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/pharmacology , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...