Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Cancer Chemother Pharmacol ; 89(1): 93-103, 2022 01.
Article in English | MEDLINE | ID: mdl-34750637

ABSTRACT

PURPOSE: New immuno-oncology therapies targeting programmed cell death receptor 1 (PD-1) have improved patient outcomes in a broad range of cancers. The objective of this analysis was to evaluate the PK, pharmacodynamics (PDy), and safety of dostarlimab monotherapy in adult patients with previously-treated advanced solid tumors who participated in parts 1 and 2A of the phase 1 GARNET study. METHODS: Part 1 featured a 3 + 3 weight-based dose-escalation study, in which 21 patients received dostarlimab 1, 3, or 10 mg/kg intravenously every 2 weeks. The 2 fixed-dose nonweight-based dosing regimens of dostarlimab 500 mg every 3 weeks (Q3W) and 1000 mg every 6 weeks (Q6W) were evaluated using a modified 6 + 6 design in part 2A (n = 13). In parts 1 and 2A, treatment with dostarlimab could continue for up to 2 years or until progression, unacceptable toxicity, patient withdrawal, investigator's decision, or death. RESULTS: The dostarlimab PK profile was dose proportional, and maximal achievable receptor occupancy (RO) was observed at all dose levels in the weight-based and fixed-dose cohorts. Trough dostarlimab concentration after administration of dostarlimab 500 mg Q3W was similar to that after dostarlimab 1000 mg Q6W, the values of which (≈40 µg/mL) projected well above the lowest dostarlimab concentration required for full peripheral RO. No dose-limiting toxicities were observed. CONCLUSIONS: Dostarlimab demonstrated consistent and predictable PK and associated PDy. The observed safety profile was acceptable and characteristic of the anti-PD-1 drug class. TRIAL REGISTRATION: ClinicalTrials.gov, NCT02715284. Registration date: March 9, 2016.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/pharmacokinetics , Neoplasms/drug therapy , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacology , Area Under Curve , Body Weight , Humans , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/pharmacology , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Treatment Outcome
2.
Immunity ; 54(6): 1154-1167.e7, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33979578

ABSTRACT

Blockade of the inhibitory receptor TIM-3 shows efficacy in cancer immunotherapy clinical trials. TIM-3 inhibits production of the chemokine CXCL9 by XCR1+ classical dendritic cells (cDC1), thereby limiting antitumor immunity in mammary carcinomas. We found that increased CXCL9 expression by splenic cDC1s upon TIM-3 blockade required type I interferons and extracellular DNA. Chemokine expression as well as combinatorial efficacy of TIM-3 blockade and paclitaxel chemotherapy were impaired by deletion of Cgas and Sting. TIM-3 blockade increased uptake of extracellular DNA by cDC1 through an endocytic process that resulted in cytoplasmic localization. DNA uptake and efficacy of TIM-3 blockade required DNA binding by HMGB1, while galectin-9-induced cell surface clustering of TIM-3 was necessary for its suppressive function. Human peripheral blood cDC1s also took up extracellular DNA upon TIM-3 blockade. Thus, TIM-3 regulates endocytosis of extracellular DNA and activation of the cytoplasmic DNA sensing cGAS-STING pathway in cDC1s, with implications for understanding the mechanisms underlying TIM-3 immunotherapy.


Subject(s)
DNA/metabolism , Dendritic Cells/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction/physiology , Animals , Biological Transport/physiology , Cell Line , Cell Line, Tumor , Chemokines/metabolism , Cytoplasm/metabolism , Endocytosis/physiology , Female , HEK293 Cells , Humans , Immunotherapy/methods , Mice , Mice, Inbred C57BL
3.
Clin Cancer Res ; 25(15): 4663-4673, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31053602

ABSTRACT

PURPOSE: To determine the tumor tissue/cell distribution, functional associations, and clinical significance of PD-1, LAG-3, and TIM-3 protein expression in human non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN: Using multiplexed quantitative immunofluorescence, we performed localized measurements of CD3, PD-1, LAG-3, and TIM-3 protein in >800 clinically annotated NSCLCs from three independent cohorts represented in tissue microarrays. Associations between the marker's expression and major genomic alterations were studied in The Cancer Genome Atlas NSCLC dataset. Using mass cytometry (CyTOF) analysis of leukocytes collected from 20 resected NSCLCs, we determined the levels, coexpression, and functional profile of PD-1, LAG-3, and TIM-3 expressing immune cells. Finally, we measured the markers in baseline samples from 90 patients with advanced NSCLC treated with PD-1 axis blockers and known response to treatment. RESULTS: PD-1, LAG-3, and TIM-3 were detected in tumor-infiltrating lymphocytes (TIL) from 55%, 41.5%, and 25.3% of NSCLC cases, respectively. These markers showed a prominent association with each other and limited association with major clinicopathologic variables and survival in patients not receiving immunotherapy. Expression of the markers was lower in EGFR-mutated adenocarcinomas and displayed limited association with tumor mutational burden. In single-cell CyTOF analysis, PD-1 and LAG-3 were predominantly localized on T-cell subsets/NKT cells, whereas TIM-3 expression was higher in NK cells and macrophages. Coexpression of PD-1, LAG-3, and TIM-3 was associated with prominent T-cell activation (CD69/CD137), effector function (Granzyme-B), and proliferation (Ki-67), but also with elevated levels of proapoptotic markers (FAS/BIM). LAG-3 and TIM-3 were present in TIL subsets lacking PD-1 expression and showed a distinct functional profile. In baseline samples from 90 patients with advanced NSCLC treated with PD-1 axis blockers, elevated LAG-3 was significantly associated with shorter progression-free survival. CONCLUSIONS: PD-1, LAG-3, and TIM-3 have distinct tissue/cell distribution, functional implications, and genomic correlates in human NSCLC. Expression of these immune inhibitory receptors in TILs is associated with prominent activation, but also with a proapoptotic T-cell phenotype. Elevated LAG-3 expression is associated with insensitivity to PD-1 axis blockade, suggesting independence of these immune evasion pathways.


Subject(s)
Antigens, CD/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Hepatitis A Virus Cellular Receptor 2/metabolism , Lung Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/metabolism , Single-Cell Analysis/methods , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lymphocyte Activation/immunology , Prognosis , Retrospective Studies , Survival Rate , Lymphocyte Activation Gene 3 Protein
4.
Br J Cancer ; 118(11): 1425-1433, 2018 05.
Article in English | MEDLINE | ID: mdl-29765150

ABSTRACT

BACKGROUND: Proviral integration Moloney virus (PIM) kinases (PIM1, 2 and 3) are overexpressed in several tumour types and contribute to oncogenesis. AZD1208 is a potent ATP-competitive PIM kinase inhibitor investigated in patients with recurrent or refractory acute myeloid leukaemia (AML) or advanced solid tumours. METHODS: Two dose-escalation studies were performed to evaluate the safety and tolerability, and to define the maximum tolerated dose (MTD), of AZD1208 in AML and solid tumours. Secondary objectives were to evaluate the pharmacokinetics, pharmacodynamics (PD) and preliminary efficacy of AZD1208. RESULTS: Sixty-seven patients received treatment: 32 in the AML study over a 120-900 mg dose range, and 25 in the solid tumour study over a 120-800 mg dose range. Nearly all patients (98.5%) in both studies experienced adverse events, mostly gastrointestinal (92.5%). Dose-limiting toxicities included rash, fatigue and vomiting. AZD1208 was not tolerated at 900 mg, and the protocol-defined MTD was not confirmed. AZD1208 increased CYP3A4 activity after multiple dosing, resulting in increased drug clearance. There were no clinical responses; PD analysis showed biological activity of AZD1208. CONCLUSIONS: Despite the lack of single-agent clinical efficacy with AZD1208, PIM kinase inhibition may hold potential as an anticancer treatment, perhaps in combination with other agents.


Subject(s)
Biphenyl Compounds/administration & dosage , Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Thiazolidines/administration & dosage , Adult , Aged , Aged, 80 and over , Biphenyl Compounds/adverse effects , Biphenyl Compounds/pharmacology , Cytochrome P-450 CYP3A/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/metabolism , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/metabolism , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacology , Thiazolidines/adverse effects , Thiazolidines/pharmacology , Up-Regulation , Young Adult
7.
Nat Med ; 22(11): 1303-1313, 2016 11.
Article in English | MEDLINE | ID: mdl-27775704

ABSTRACT

Triple-negative breast cancers (TNBCs) have poor prognosis and lack targeted therapies. Here we identified increased copy number and expression of the PIM1 proto-oncogene in genomic data sets of patients with TNBC. TNBC cells, but not nonmalignant mammary epithelial cells, were dependent on PIM1 for proliferation and protection from apoptosis. PIM1 knockdown reduced expression of the anti-apoptotic factor BCL2, and dynamic BH3 profiling of apoptotic priming revealed that PIM1 prevents mitochondrial-mediated apoptosis in TNBC cell lines. In TNBC tumors and their cellular models, PIM1 expression was associated with several transcriptional signatures involving the transcription factor MYC, and PIM1 depletion in TNBC cell lines decreased, in a MYC-dependent manner, cell population growth and expression of the MYC target gene MCL1. Treatment with the pan-PIM kinase inhibitor AZD1208 impaired the growth of both cell line and patient-derived xenografts and sensitized them to standard-of-care chemotherapy. This work identifies PIM1 as a malignant-cell-selective target in TNBC and the potential use of PIM1 inhibitors for sensitizing TNBC to chemotherapy-induced apoptotic cell death.


Subject(s)
Apoptosis/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-pim-1/genetics , Triple Negative Breast Neoplasms/genetics , Animals , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , DNA Copy Number Variations , Female , Gene Knockdown Techniques , Humans , Mice , Mitochondria/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasm Transplantation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Real-Time Polymerase Chain Reaction , Thiazolidines/pharmacology , Xenograft Model Antitumor Assays
8.
Mol Cell Biol ; 34(13): 2517-32, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24777602

ABSTRACT

MET, the receptor for hepatocyte growth factor (HGF), plays an important role in signaling normal and tumor cell migration and invasion. Here, we describe a previously unrecognized mechanism that promotes MET expression in multiple tumor cell types. The levels of the Pim-1 protein kinase show a positive correlation with the levels of MET protein in human tumor cell lines and patient-derived tumor materials. Using small interfering RNA (siRNA), Pim knockout mice, small-molecule inhibitors, and overexpression of Pim-1, we confirmed this correlation and found that Pim-1 kinase activity regulates HGF-induced tumor cell migration, invasion, and cell scattering. The novel biochemical mechanism for these effects involves the ability of Pim-1 to control the translation of MET by regulating the phosphorylation of eukaryotic initiation factor 4B (eIF4B) on S406. This targeted phosphorylation is required for the binding of eIF4B to the eIF3 translation initiation complex. Importantly, Pim-1 action was validated by the evaluation of patient blood and bone marrow from a phase I clinical trial of a Pim kinase inhibitor, AZD1208. These results suggest that Pim inhibitors may have an important role in the treatment of patients where MET is driving tumor biology.


Subject(s)
Eukaryotic Initiation Factors/genetics , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins c-met/biosynthesis , Proto-Oncogene Proteins c-pim-1/biosynthesis , Animals , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Cell Movement , Eukaryotic Initiation Factor-3/genetics , HeLa Cells , Humans , Mice , Mice, Knockout , Neoplasm Invasiveness , Phosphorylation , Protein Binding , Protein Biosynthesis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/genetics , RNA Interference , RNA, Small Interfering , Signal Transduction , Thiazolidines/pharmacology
9.
Blood ; 123(6): 905-13, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24363397

ABSTRACT

Upregulation of Pim kinases is observed in several types of leukemias and lymphomas. Pim-1, -2, and -3 promote cell proliferation and survival downstream of cytokine and growth factor signaling pathways. AZD1208 is a potent, highly selective, and orally available Pim kinase inhibitor that effectively inhibits all three isoforms at <5 nM or <150 nM in enzyme and cell assays, respectively. AZD1208 inhibited the growth of 5 of 14 acute myeloid leukemia (AML) cell lines tested, and sensitivity correlates with Pim-1 expression and STAT5 activation. AZD1208 causes cell cycle arrest and apoptosis in MOLM-16 cells, accompanied by a dose-dependent reduction in phosphorylation of Bcl-2 antagonist of cell death, 4EBP1, p70S6K, and S6, as well as increases in cleaved caspase 3 and p27. Inhibition of p4EBP1 and p-p70S6K and suppression of translation are the most representative effects of Pim inhibition in sensitive AML cell lines. AZD1208 inhibits the growth of MOLM-16 and KG-1a xenograft tumors in vivo with a clear pharmacodynamic-pharmacokinetic relationship. AZD1208 also potently inhibits colony growth and Pim signaling substrates in primary AML cells from bone marrow that are Flt3 wild-type or Flt3 internal tandem duplication mutant. These results underscore the therapeutic potential of Pim kinase inhibition for the treatment of AML.


Subject(s)
Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Cell Proliferation/drug effects , Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Thiazolidines/pharmacology , Animals , Biphenyl Compounds/pharmacokinetics , Blotting, Western , Cell Cycle , Enzyme-Linked Immunosorbent Assay , Female , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Mice , Mice, SCID , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-pim-1/metabolism , Thiazolidines/pharmacokinetics , Tissue Distribution , Tumor Cells, Cultured
10.
Mol Cancer Ther ; 13(2): 386-98, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24344235

ABSTRACT

The hedgehog pathway has been implicated in the tumorigenesis, tumor progression, and metastasis of numerous human cancers. We generated the first fully human hedgehog antibody MEDI-5304 and characterized its antitumor activity and preclinical toxicology. MEDI-5304 bound sonic hedgehog (SHH) and Indian hedgehog (IHH) with low picomolar affinity and neutralized SHH and IHH activity in cellular mGLI1 reporter assays. The antibody inhibited transcription of hedgehog target genes and osteoblast differentiation of C3H10T1/2 cells. We evaluated the activity of MEDI-5304 in vivo in model systems that allowed us to evaluate two primary hypotheses of hedgehog function in human cancer, paracrine signaling between tumor and stromal cells and cancer stem cell (CSC) self-renewal. MEDI-5304 displayed robust pharmacodynamic effects in stromal cells that translated to antitumor efficacy as a single agent in an HT-29/MEF coimplantation model of paracrine hedgehog signaling. MEDI-5304 also improved responses to carboplatin in the HT-29/MEF model. The antibody, however, had no effect as a single agent or in combination with gemcitabine on the CSC frequency or growth of several primary pancreatic cancer explant models. These findings support the conclusion that hedgehog contributes to tumor biology via paracrine tumor-stromal signaling but not via CSC maintenance or propagation. Finally, the only safety study finding associated with MEDI-5304 was ondontodysplasia in rats. Thus, MEDI-5304 represents a potent dual hedgehog inhibitor suitable for continued development to evaluate efficacy and safety in human patients with tumors harboring elevated levels of SHH or IHH.


Subject(s)
Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Paracrine Communication/drug effects , Animals , Antibodies, Monoclonal, Humanized , Antibodies, Neutralizing/immunology , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacokinetics , Cell Line , Cell Line, Tumor , Cells, Cultured , Female , HT29 Cells , Hedgehog Proteins/immunology , Humans , Kinetics , Macaca fascicularis , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , NIH 3T3 Cells , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Paracrine Communication/immunology , Protein Binding/immunology , Rats, Wistar , Stromal Cells/drug effects , Stromal Cells/immunology , Stromal Cells/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
11.
Cancer Cell ; 16(6): 487-97, 2009 Dec 08.
Article in English | MEDLINE | ID: mdl-19962667

ABSTRACT

Persistent activation of Stat3 is oncogenic and is prevalent in a wide variety of human cancers. Chronic cytokine stimulation is associated with Stat3 activation in some tumors, implicating cytokine receptor-associated Jak family kinases. Using Jak2 inhibitors, we demonstrate a central role of Jaks in modulating basal and cytokine-induced Stat3 activation in human solid tumor cell lines. Inhibition of Jak2 activity is associated with abrogation of Stat3 nuclear translocation and tumorigenesis. The Jak2 inhibitor AZD1480 suppresses the growth of human solid tumor xenografts harboring persistent Stat3 activity. We demonstrate the essential role of Stat3 downstream of Jaks by inhibition of tumor growth using short hairpin RNA targeting Stat3. Our data support a key role of Jak kinase activity in Stat3-dependent tumorigenesis.


Subject(s)
Janus Kinase 2/antagonists & inhibitors , Prostatic Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Cell Line, Tumor , Humans , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , STAT3 Transcription Factor/metabolism
12.
J Cell Biochem ; 89(1): 120-32, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12682913

ABSTRACT

The global changes in gene expression induced by transient increased expression of full length BRCA1 as well as the spliced variant BRCA1(S) were evaluated by cDNA expression array in a human non-tumorigenic mammary epithelial cell line, MCF10A. Over 30 genes were identified that displayed an altered expression pattern in response to the expression of BRCA1 splice variants. The expression of NFkappaB inducing kinase was markedly down-regulated in BRCA1(L) transfected cells. However, a NFkappaB-responsive promoter construct yielded increased basal activity in BRCA1(L) transfected cells, as well as following treatment with tumor necrosis factor-alpha or lymphotoxin. In addition, nuclear extracts from BRCA1(L) transfected cells displayed increased DNA binding to the kappaB consensus site. The transcriptional activity of a panel of promoter constructs was evaluated following expression of wild type or mutant BRCA1. Full length BRCA1 transactivated the estrogen receptor-alpha (ERalpha) and BCL2 promoters as well as AP-1, SRE, and CRE containing promoters. Transactivation activity of the exon 11-deleted BRCA1(S) was more limited and usually of lower magnitude. The ability of a pathogenic mutation, 5382insC, to abrogate the transcriptional transactivation by BRCA1(L) and BRCA1(S) was also investigated. Mutant BRCA1 retained wild type levels of transcriptional activity for the ERalpha promoter as well as for the NFkappaB, AP-1, and CRE-responsive promoters but had reduced or no activity with the BCL2 and SRE promoters. These results show that BRCA1 isoforms have both overlapping and distinct transcriptional transactivation activity, and that a mutant form of BRCA1 implicated in carcinogenesis is not devoid of all activity.


Subject(s)
Alternative Splicing , Genes, BRCA1 , Base Sequence , Breast/cytology , Breast/metabolism , Cell Line , DNA, Complementary/genetics , Female , Gene Expression , Humans , NF-kappa B/metabolism , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Transcriptional Activation , Transfection , NF-kappaB-Inducing Kinase
13.
Oncogene ; 21(46): 7034-41, 2002 Oct 10.
Article in English | MEDLINE | ID: mdl-12370825

ABSTRACT

A distinctive feature of BRCA1-linked breast cancers is that they typically do not express estrogen receptor-alpha (ER(alpha)). Previous investigation suggests that methylation of CpGs within the ER(alpha) promoter mediates repression of gene expression in some ER(alpha)-negative breast cancers. To determine if methylation of CpGs within the ER(alpha) promoter is associated with BRCA1-linked breast cancers, we evaluated methylation in exon 1 of the ER(alpha) gene in 40 ER(alpha)-negative breast cancers, 20 of which were non BRCA1-linked and 20 BRCA1-linked. CpG methylation was evaluated by either methylation-sensitive restriction digest (HpaII), methylation-sensitive PCR (MSP), or direct sequencing of bisulfite-treated genomic DNA. Results from HpaII digests and MSP documented a high degree of methylation, the MSP data showing slightly higher methylation in the BRCA1-linked group. CpGs analysed by direct sequencing showed an overall average methylation of 25% among non BRCA1-linked cancers and 40% among BRCA1-linked cancers (P=0.0031). The most notable difference was found at five particular CpGs, each of which exhibited a greater than twofold increase in methylation in the BRCA1-linked group compared to the non BRCA1-linked group (P<0.03 for each CpG). Methylation of certain critical CpGs may represent an important factor in transcriptional repression of the ER(alpha) gene in BRCA1-linked breast cancers.


Subject(s)
Breast Neoplasms/genetics , CpG Islands , DNA Methylation , Genes, BRCA1 , Receptors, Estrogen/genetics , Adult , Aged , Breast Neoplasms/chemistry , Estrogen Receptor alpha , Female , Genetic Linkage , Humans , Middle Aged , Polymerase Chain Reaction , Promoter Regions, Genetic , Receptors, Estrogen/analysis , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...