Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Breast Care (Basel) ; 19(1): 49-61, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38384487

ABSTRACT

Background: We investigated the efficacy and health-related quality of life (HRQoL) in patients receiving either ribociclib plus endocrine therapy (ET) or chemotherapy with/without bevacizumab as first-line treatment of metastatic hormone receptor (HR)-positive, HER2-negative breast cancer (BC). Patients and Methods: In this randomized, phase III study (RIBBIT), 38 patients diagnosed with metastatic HR-positive, HER2-negative BC with presence of visceral metastases recruited between May 2018 and December 2020 were randomly assigned in a 1:1 ratio to either arm A (ribociclib + ET) or arm B (chemotherapy with/without bevacizumab) at 12 sites in Germany. The primary endpoint was progression-free survival (PFS). Secondary endpoints included overall response rate (ORR), overall survival (OS), patient-reported HRQoL, and frequency and type of adverse events. During study conduction, the recruitment rate was persistently and considerably lower than originally expected. Therefore, the recruitment was ended prematurely. The study was initially designed to enroll and randomize 158 patients. Results: Median [95% CI] PFS was 27.3 months [19.1 - NA, parameter not estimable] in arm A and 15.8 months [8.2 - NA] in arm B. Complete responses were achieved only in arm A (n = 2, 10.5%). The ORR [95% CI] between arm A (57.9% [33.5-79.7]) and arm B (52.6% [28.9-75.6]) was comparable. Median OS [95% CI] was not reached in arm A, while in arm B median OS was 28.4 months [25.0 - NA]. Patients in arm A reported less burden by side-effects. No new safety signals emerged. Conclusion: Treatment of patients with visceral metastatic HR-positive, HER2-negative BC with ribociclib in combination with ET showed a tendency toward a more favorable clinical outcome. Despite small numbers of patients and sites, this head-to-head comparison with chemotherapy supports the use of ribociclib with ET in patients with visceral metastasis at risk of fast disease progression.

2.
BMC Cancer ; 23(1): 543, 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37312086

ABSTRACT

BACKGROUND: Precision oncology, defined as treatment of patients with targeted therapies matched to specific molecular alterations, has entered routine clinical practice. Particularly in patients with advanced cancer or hematologic malignancies, for whom no further standard therapies are available, this approach is increasingly applied as last resort option outside of the approved indication. However, data on patient outcomes are not systematically collected, analyzed, reported, and shared. We have initiated the INFINITY registry to provide evidence from routine clinical practice to fill this knowledge gap. METHODS: INFINITY is a retrospective, non-interventional cohort study conducted at approximately 100 sites in Germany (office-based oncologists/hematologists and hospitals). We aim to include 500 patients with advanced solid tumors or hematologic malignancies who received a non-standard targeted therapy based on potentially actionable molecular alterations or biomarkers. INFINITY aims to provide insights into the use of precision oncology in routine clinical practice within Germany. We systematically collect details on patient and disease characteristics, molecular testing, clinical decision-making, treatment, and outcome. DISCUSSION: INFINITY will provide evidence on the current biomarker landscape driving treatment decisions in routine clinical care. It will also provide insights on effectiveness of precision oncology approaches in general, and of specific drug class/alteration matches used outside their approved indications. TRIAL REGISTRATION: The study is registered at ClinicalTrials.gov, NCT04389541.


Subject(s)
Hematologic Neoplasms , Neoplasms , Humans , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/genetics , Retrospective Studies , Cohort Studies , Precision Medicine , Biomarkers , Decision Making
3.
Cancer Immunol Immunother ; 66(12): 1609-1617, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28856392

ABSTRACT

The ability of some tumors to exclude effector T cells represents a major challenge to immunotherapy. T cell exclusion is particularly evident in pancreatic ductal adenocarcinoma (PDAC), a disease where blockade of the immune checkpoint molecule CTLA-4 has not produced significant clinical activity. In PDAC, effector T cells are often scarce within tumor tissue and confined to peritumoral lymph nodes and lymphoid aggregates. We hypothesized that CTLA-4 blockade, despite a lack of clinical efficacy seen thus far in PDAC, might still alter T cell immunobiology, which would have therapeutic implications. Using clinically relevant genetic models of PDAC, we found that regulatory T cells (Tregs), which constitutively express CTLA-4, accumulate early during tumor development but are largely confined to peritumoral lymph nodes during disease progression. Tregs were observed to regulate CD4+, but not CD8+, T cell infiltration into tumors through a CTLA-4/CD80 dependent mechanism. Disrupting CTLA-4 interaction with CD80 was sufficient to induce CD4 T cell infiltration into tumors. These data have important implications for T cell immunotherapy in PDAC and demonstrate a novel role for CTLA-4/CD80 interactions in regulating T cell exclusion. In addition, our findings suggest distinct mechanisms govern CD4+ and CD8+ T cell infiltration in PDAC.


Subject(s)
B7-1 Antigen/immunology , CTLA-4 Antigen/immunology , Pancreatic Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies/immunology , Antibodies/pharmacology , CTLA-4 Antigen/antagonists & inhibitors , Humans , Interleukin-2 Receptor alpha Subunit/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Signal Transduction , Pancreatic Neoplasms
4.
Cell Rep ; 16(11): 2829-2837, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27626654

ABSTRACT

Restoration of anti-tumor immunity by blocking PD-L1 signaling through the use of antibodies has proven to be beneficial in cancer therapy. Here, we show that BET bromodomain inhibition suppresses PD-L1 expression and limits tumor progression in ovarian cancer. CD274 (encoding PD-L1) is a direct target of BRD4-mediated gene transcription. In mouse models, treatment with the BET inhibitor JQ1 significantly reduced PD-L1 expression on tumor cells and tumor-associated dendritic cells and macrophages, which correlated with an increase in the activity of anti-tumor cytotoxic T cells. The BET inhibitor limited tumor progression in a cytotoxic T-cell-dependent manner. Together, these data demonstrate a small-molecule approach to block PD-L1 signaling. Given the fact that BET inhibitors have been proven to be safe with manageable reversible toxicity in clinical trials, our findings indicate that pharmacological BET inhibitors represent a treatment strategy for targeting PD-L1 expression.


Subject(s)
Azepines/pharmacology , B7-H1 Antigen/genetics , Immunity/drug effects , Neoplasms/immunology , Neoplasms/metabolism , Proteins/antagonists & inhibitors , Triazoles/pharmacology , Animals , B7-H1 Antigen/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Disease Progression , Female , Humans , Mice, Inbred C57BL , Neoplasms/pathology , Nuclear Proteins/metabolism , Proteins/metabolism , T-Lymphocytes, Cytotoxic/drug effects , Time Factors , Transcription Factors/metabolism , Transcription, Genetic/drug effects
5.
Oncotarget ; 7(36): 58244-58260, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27542270

ABSTRACT

Tumor initiating cells (TICs) have been identified and functionally characterized in hematological malignancies as well as in solid tumors such as breast cancer. In addition to their high tumor-initiating potential, TICs are founder cells for metastasis formation and are involved in chemotherapy resistance. In this study we explored molecular pathways which enable this tumor initiating potential for a cancer cell subset of the transgenic MMTV-PyMT mouse model for metastasizing breast cancer. The cell population, characterized by the marker profile CD24+CD90+CD45-, showed a high tumorigenicity compared to non-CD24+CD90+CD45- cancer cells in colony formation assays, as well as upon orthotopic transplantation into the mammary fat pad of mice. In addition, these orthotopically grown CD24+CD90+CD45- TICs metastasized to the lungs. The transcriptome of TICs freshly isolated from primary tumors by cell sorting was compared with that of sorted non-CD24+CD90+CD45- cancer cells by RNA-seq. In addition to more established TIC signatures, such as epithelial-to-mesenchymal transition or mitogen signaling, an upregulated gene set comprising several classes of proteolytic enzymes was uncovered in the TICs. Accordingly, TICs showed high intra- and extracellular proteolytic activity. Application of a broad range of protease inhibitors to TICs in a colony formation assay reduced anchorage independent growth and had an impact on colony morphology in 3D cell culture assays. We conclude that CD24+CD90+CD45- cells of the MMTV- PyMT mouse model possess an upregulated proteolytic signature which could very well represent a functional hallmark of metastatic TICs from mammary carcinomas.


Subject(s)
Mammary Neoplasms, Animal/pathology , Neoplastic Stem Cells/pathology , Proteolysis , Animals , CD24 Antigen/metabolism , Carcinogenesis , Cell Culture Techniques , Cell Proliferation , Cell Separation , Cell Transformation, Neoplastic/metabolism , Female , Flow Cytometry , Gene Expression Regulation , Leukocyte Common Antigens/metabolism , Mammary Neoplasms, Animal/therapy , Matrix Metalloproteinase 14/metabolism , Mice , Mice, Transgenic , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Peptide Hydrolases/metabolism , Sequence Analysis, RNA , Signal Transduction , Thy-1 Antigens/metabolism , Transcriptome
6.
Curr Protoc Pharmacol ; 73: 14.39.1-14.39.20, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27248578

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) ranks fourth among cancer-related deaths in the United States. For patients with unresectable disease, treatment options are limited and lack curative potential. Preclinical mouse models of PDAC that recapitulate the biology of human pancreatic cancer offer an opportunity for the rational development of novel treatment approaches that may improve patient outcomes. With the recent success of immunotherapy for subsets of patients with solid malignancies, interest is mounting in the possible use of immunotherapy for the treatment of PDAC. Considered in this unit is the value of genetic mouse models for characterizing the immunobiology of PDAC and for investigating novel immunotherapeutics. Several variants of these models are described, all of which may be used in drug development and for providing information on unique aspects of disease biology and therapeutic responsiveness. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Animals, Genetically Modified , Carcinoma, Pancreatic Ductal , Disease Models, Animal , Mice , Pancreatic Neoplasms , Animals , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/therapy , Drug Discovery , Humans , Immunotherapy , Macrophages , Mice/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , T-Lymphocytes
7.
Cell Cycle ; 14(10): 1540-7, 2015.
Article in English | MEDLINE | ID: mdl-25785348

ABSTRACT

Oncogene-induced senescence (OIS) is an important tumor suppression mechanism preventing uncontrolled proliferation in response to aberrant oncogenic signaling. The profound functional and morphological remodelling of the senescent cell involves extensive changes. In particular, alterations in protein ubiquitination during senescence have not been systematically analyzed previously. Here, we report the first global ubiquitination profile of primary human cells undergoing senescence. We employed a well-characterized in vitro model of OIS, primary human fibroblasts expressing oncogenic RAS. To compare the ubiquitinome of RAS-induced OIS and controls, ubiquitinated peptides were enriched by immune affinity purification and subjected to liquid chromatography tandem mass spectrometry (LC-MS/MS). We identified 4,472 ubiquitination sites, with 397 sites significantly changed (>3 standard deviations) in senescent cells. In addition, we performed mass spectrometry analysis of total proteins in OIS and control cells to account for parallel changes in both protein abundance and ubiquitin levels that did not affect the percentage of ubiquitination of a given protein. Pathway analysis revealed that the OIS-induced ubiquitinome alterations mainly affected 3 signaling networks: eIF2 signaling, eIF4/p70S6K signaling, and mTOR signaling. Interestingly, the majority of the changed ubiquitinated proteins in these pathways belong to the translation machinery. This includes several translation initiation factors (eIF2C2, eIF2B4, eIF3I, eIF3L, eIF4A1) and elongation factors (eEF1G, eEF1A) as well as 40S (RPS4X, RPS7, RPS11 and RPS20) and 60S ribosomal subunits (RPL10, RPL11, RPL18 and RPL35a). In addition, we observed enriched ubiquitination of aminoacyl-tRNA ligases (isoleucyl-, glutamine-, and tyrosine-tRNA ligase), which provide the amino acid-loaded tRNAs for protein synthesis. These results suggest that ubiquitination affects key components of the translation machinery to regulate protein synthesis during OIS. Our results thus point toward ubiquitination as a hitherto unappreciated regulatory mechanism during OIS.


Subject(s)
Cellular Senescence , Oncogene Protein p21(ras)/metabolism , Amino Acid Sequence , Cells, Cultured , Chromatography, High Pressure Liquid , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-4F/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Isotope Labeling , Oncogene Protein p21(ras)/genetics , Peptides/analysis , Peptides/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tandem Mass Spectrometry , Ubiquitination
8.
Mol Cell Proteomics ; 12(3): 611-25, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23233448

ABSTRACT

Numerous studies highlight the fact that concerted proteolysis is essential for skin morphology and function. The cysteine protease cathepsin L (Ctsl) has been implicated in epidermal proliferation and desquamation, as well as in hair cycle regulation. In stark contrast, mice deficient in cathepsin B (Ctsb) do not display an overt skin phenotype. To understand the systematic consequences of deleting Ctsb or Ctsl, we determined the protein abundances of >1300 proteins and proteolytic cleavage events in skin samples of wild-type, Ctsb(-/-), and Ctsl(-/-) mice via mass-spectrometry-based proteomics. Both protease deficiencies revealed distinct quantitative changes in proteome composition. Ctsl(-/-) skin revealed increased levels of the cysteine protease inhibitors cystatin B and cystatin M/E, increased cathepsin D, and an accumulation of the extracellular glycoprotein periostin. Immunohistochemistry located periostin predominantly in the hypodermal connective tissue of Ctsl(-/-) skin. The proteomic identification of proteolytic cleavage sites within skin proteins revealed numerous processing sites that are underrepresented in Ctsl(-/-) or Ctsb(-/-) samples. Notably, few of the affected cleavage sites shared the canonical Ctsl or Ctsb specificity, providing further evidence of a complex proteolytic network in the skin. Novel processing sites in proteins such as dermokine and Notch-1 were detected. Simultaneous analysis of acetylated protein N termini showed prototypical mammalian N-alpha acetylation. These results illustrate an influence of both Ctsb and Ctsl on the murine skin proteome and degradome, with the phenotypic consequences of the absence of either protease differing considerably.


Subject(s)
Cathepsin B/deficiency , Cathepsin L/deficiency , Proteome/metabolism , Proteomics/methods , Skin/metabolism , Animals , Blotting, Western , Cathepsin B/genetics , Cathepsin L/genetics , Cell Adhesion Molecules/metabolism , Cells, Cultured , Chromatography, Liquid , Cystatin B/metabolism , Cystatin M/metabolism , Immunohistochemistry , Mice , Mice, Knockout , Peptides/metabolism , Proteolysis , Receptor, Notch1/metabolism , Serpins/metabolism , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...