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










Publication year range
1.
Ann Hematol ; 103(6): 1989-2001, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38233570

ABSTRACT

Natural killer/T cell lymphoma (NKTCL) is a highly aggressive, heterogeneous non-Hodgkin lymphoma resulting from malignant proliferation of cytotoxic natural killer (NK) or T cells. Previous studies demonstrated variable expression of CD38 on NKTCL tumors. Daratumumab, a human IgGκ monoclonal antibody targeting CD38 with a direct on-tumor and immunomodulatory mechanism of action, was hypothesized to be a novel therapeutic option for patients with relapsed or refractory (R/R) NKTCL. In the phase 2 NKT2001 study (ClinicalTrials.gov Identifier: NCT02927925) assessing the safety and efficacy of daratumumab, a suboptimal overall response rate was seen in R/R NKTCL patients. One patient, whose tumors did not express CD38, responded to treatment, suggesting that the immunomodulatory activities of daratumumab may be sufficient to confer clinical benefit. To understand the suboptimal response rate and short duration of response, we investigated the immune profile of NKTCL patients from NKT2001 in the context of daratumumab anti-tumor activity. Tumor tissue and whole blood were, respectively, analyzed for CD38 expression and patient immune landscapes, which were assessed via cytometry by time-of-flight (CyTOF), multiparameter flow cytometry (MPFC), clonal sequencing, and plasma Epstein-Barr virus (EBV)-DNA level measurements. Changes observed in the immune profiles of NKTCL patients from NKT2001, including differences in B and T cell populations between responders and nonresponders, suggest that modulation of the immune environment is crucial for daratumumab anti-tumor activities in NKTCL. In conclusion, these findings highlight that the clinical benefit of daratumumab in NKTCL may be enriched by B/T cell-related biomarkers.


Subject(s)
Antibodies, Monoclonal , Lymphoma, Extranodal NK-T-Cell , Humans , Antibodies, Monoclonal/therapeutic use , Lymphoma, Extranodal NK-T-Cell/drug therapy , Lymphoma, Extranodal NK-T-Cell/immunology , Male , Female , ADP-ribosyl Cyclase 1 , Middle Aged , Aged , Adult , Membrane Glycoproteins
2.
Clin Transl Med ; 13(12): e1507, 2023 12.
Article in English | MEDLINE | ID: mdl-38115705

ABSTRACT

Whereas most infants infected with respiratory syncytial virus (RSV) show no or only mild symptoms, an estimated 3 million children under five are hospitalized annually due to RSV disease. This study aimed to investigate biological mechanisms and associated biomarkers underlying RSV disease heterogeneity in young infants, enabling the potential to objectively categorize RSV-infected infants according to their medical needs. Immunophenotypic and functional profiling demonstrated the emergence of immature and progenitor-like neutrophils, proliferative monocytes (HLA-DRLow , Ki67+), impaired antigen-presenting function, downregulation of T cell response and low abundance of HLA-DRLow B cells in severe RSV disease. HLA-DRLow monocytes were found as a hallmark of RSV-infected infants requiring hospitalization. Complementary transcriptomics identified genes associated with disease severity and pointed to the emergency myelopoiesis response. These results shed new light on mechanisms underlying the pathogenesis and development of severe RSV disease and identified potential new candidate biomarkers for patient stratification.


Subject(s)
Myelopoiesis , Respiratory Syncytial Virus Infections , Infant , Child , Humans , Myelopoiesis/genetics , Respiratory Syncytial Virus Infections/diagnosis , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Viruses , HLA-DR Antigens , Biomarkers
3.
Hemasphere ; 7(5): e881, 2023 May.
Article in English | MEDLINE | ID: mdl-37153876

ABSTRACT

The CD38-targeting antibody daratumumab has marked activity in multiple myeloma (MM). Natural killer (NK) cells play an important role during daratumumab therapy by mediating antibody-dependent cellular cytotoxicity via their FcγRIII receptor (CD16), but they are also rapidly decreased following initiation of daratumumab treatment. We characterized the NK cell phenotype at baseline and during daratumumab monotherapy by flow cytometry and cytometry by time of flight to assess its impact on response and development of resistance (DARA-ATRA study; NCT02751255). At baseline, nonresponding patients had a significantly lower proportion of CD16+ and granzyme B+ NK cells, and higher frequency of TIM-3+ and HLA-DR+ NK cells, consistent with a more activated/exhausted phenotype. These NK cell characteristics were also predictive of inferior progression-free survival and overall survival. Upon initiation of daratumumab treatment, NK cells were rapidly depleted. Persisting NK cells exhibited an activated and exhausted phenotype with reduced expression of CD16 and granzyme B, and increased expression of TIM-3 and HLA-DR. We observed that addition of healthy donor-derived purified NK cells to BM samples from patients with either primary or acquired daratumumab-resistance improved daratumumab-mediated MM cell killing. In conclusion, NK cell dysfunction plays a role in primary and acquired daratumumab resistance. This study supports the clinical evaluation of daratumumab combined with adoptive transfer of NK cells.

4.
Leukemia ; 35(2): 573-584, 2021 02.
Article in English | MEDLINE | ID: mdl-32457357

ABSTRACT

CD38-targeted antibody, daratumumab, is approved for the treatment of multiple myeloma (MM). Phase 1/2 studies GEN501/SIRIUS revealed a novel immunomodulatory mechanism of action (MOA) of daratumumab that enhanced the immune response, reducing natural killer (NK) cells without affecting efficacy or safety. We further evaluated daratumumab's effects on immune cells in whole blood samples of relapsed/refractory MM patients from both treatment arms of the phase 3 POLLUX study (lenalidomide/dexamethasone [Rd] or daratumumab plus Rd [D-Rd]) at baseline (D-Rd, 40; Rd, 45) and after 2 months on treatment (D-Rd, 31; Rd, 33) using cytometry by time-of-flight. We confirmed previous reports of NK cell reduction with D-Rd. Persisting NK cells were phenotypically distinct, with increased expression of HLA-DR, CD69, CD127, and CD27. The proportion of T cells increased preferentially in deep responders to D-Rd, with a higher proportion of CD8+ versus CD4+ T cells. The expansion of CD8+ T cells correlated with clonality, indicating generation of adaptive immune response with D-Rd. D-Rd resulted in a higher proportion of effector memory T cells versus Rd. D-Rd reduced immunosuppressive CD38+ regulatory T cells. This study confirms daratumumab's immunomodulatory MOA in combination with immunomodulatory drugs and provides further insight into immune cell changes and activation status following daratumumab-based therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers/analysis , Killer Cells, Natural/immunology , Multiple Myeloma/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes/immunology , Antibodies, Monoclonal/administration & dosage , Dexamethasone/administration & dosage , Humans , Killer Cells, Natural/drug effects , Lenalidomide/administration & dosage , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , T-Lymphocytes/drug effects , T-Lymphocytes, Regulatory/drug effects
5.
Cytometry A ; 95(3): 279-289, 2019 03.
Article in English | MEDLINE | ID: mdl-30536810

ABSTRACT

Daratumumab is a CD38-targeted human monoclonal antibody with direct anti-myeloma cell mechanisms of action. Flow cytometry in relapsed and/or refractory multiple myeloma (RRMM) patients treated with daratumumab revealed cytotoxic T-cell expansion and reduction of immune-suppressive populations, suggesting immune modulation as an additional mechanism of action. Here, we performed an in-depth analysis of the effects of daratumumab on immune-cell subpopulations using high-dimensional mass cytometry. Whole-blood and bone-marrow baseline and on-treatment samples from RRMM patients who participated in daratumumab monotherapy studies (SIRIUS and GEN501) were evaluated with high-throughput immunophenotyping. In daratumumab-treated patients, the intensity of CD38 marker expression decreased on many immune cells in SIRIUS whole-blood samples. Natural killer (NK) cells were depleted with daratumumab, with remaining NK cells showing increased CD69 and CD127, decreased CD45RA, and trends for increased CD25, CD27, and CD137 and decreased granzyme B. Immune-suppressive population depletion paralleled previous findings, and a newly observed reduction in CD38+ basophils was seen in patients who received monotherapy. After 2 months of daratumumab, the T-cell population in whole-blood samples from responders shifted to a CD8 prevalence with higher granzyme B positivity (P = 0.017), suggesting increased killing capacity and supporting monotherapy-induced CD8+ T-cell activation. High-throughput cytometry immune profiling confirms and builds upon previous flow cytometry data, including comparable CD38 marker intensity on plasma cells, NK cells, monocytes, and B/T cells. Interestingly, a shift toward cytolytic granzyme B+ T cells was also observed and supports adaptive responses in patients that may contribute to depth of response. © 2018 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , Basophils/cytology , Basophils/drug effects , Basophils/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Flow Cytometry , Granzymes/metabolism , Humans , Immunophenotyping , Killer Cells, Natural/cytology , Multiple Myeloma/blood , Multiple Myeloma/metabolism , Recurrence
6.
Cytometry B Clin Cytom ; 92(1): 42-56, 2017 01.
Article in English | MEDLINE | ID: mdl-27933748

ABSTRACT

Recent advances in combining flow cytometry and mass spectrometry have led to the development of mass cytometry, allowing for the interrogation of complex cell populations on an unprecedented scale. The volumes and high dimensionality of mass cytometry data pose significant challenges in terms of analysis and visualization. We implement a method called Radviz, where multidimensional single cell data can be visualized as a projection that maintains the original dimensions and data complexity whilst facilitating analysis and visualization. This enables identification of changes in populations, focusing the analysis on the most relevant aspect of large multidimensional datasets. To highlight the potential of Radviz, we profiled peripheral mononuclear blood cells (PBMCs) from three healthy donors and showed donor-specific differences in the number and composition of cell populations. In a second study, we explored the anti-inflammatory effects of two glucocorticoid receptor (GR) ligands (cpd6 and cpd11) compared to dexamethasone (Dex) on human primary macrophages. Standard analysis at the population level showed that cpd6 and cpd11 have an overall anti-inflammatory profile similar to that of Dex. CyTOF profiling and Radviz-driven analysis at the single cell level confirmed this observation, and identified a concentration-dependent effect of cpd6 that was not detected at the population level. Altogether, Radviz combines the strengths of a projection method, reducing the dimensionality of datasets, with that of a scatter plot, where the identity of each point can be inferred from the distance to the axis. This enables the visual exploration, analysis, and interpretation of complex, high dimensional data. © 2016 International Clinical Cytometry Society.


Subject(s)
Dexamethasone/pharmacology , Flow Cytometry , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/immunology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cell Line , Flow Cytometry/methods , Humans , Leukocytes, Mononuclear/drug effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Receptors, Cell Surface/metabolism
7.
SLAS Discov ; 22(3): 238-249, 2017 03.
Article in English | MEDLINE | ID: mdl-27899692

ABSTRACT

High-throughput screening generates large volumes of heterogeneous data that require a diverse set of computational tools for management, processing, and analysis. Building integrated, scalable, and robust computational workflows for such applications is challenging but highly valuable. Scientific data integration and pipelining facilitate standardized data processing, collaboration, and reuse of best practices. We describe how Jenkins-CI, an "off-the-shelf," open-source, continuous integration system, is used to build pipelines for processing images and associated data from high-content screening (HCS). Jenkins-CI provides numerous plugins for standard compute tasks, and its design allows the quick integration of external scientific applications. Using Jenkins-CI, we integrated CellProfiler, an open-source image-processing platform, with various HCS utilities and a high-performance Linux cluster. The platform is web-accessible, facilitates access and sharing of high-performance compute resources, and automates previously cumbersome data and image-processing tasks. Imaging pipelines developed using the desktop CellProfiler client can be managed and shared through a centralized Jenkins-CI repository. Pipelines and managed data are annotated to facilitate collaboration and reuse. Limitations with Jenkins-CI (primarily around the user interface) were addressed through the selection of helper plugins from the Jenkins-CI community.


Subject(s)
Algorithms , Image Processing, Computer-Assisted/statistics & numerical data , Molecular Imaging/statistics & numerical data , User-Computer Interface , Animals , Cell Line , Gene Expression Regulation , Humans , Internet , Molecular Imaging/methods , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Workflow
8.
Hepatology ; 62(5): 1497-510, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26173433

ABSTRACT

UNLABELLED: The Yes-associated protein (YAP)/Hippo pathway has been implicated in tissue development, regeneration, and tumorigenesis. However, its role in cholangiocarcinoma (CC) is not established. We show that YAP activation is a common feature in CC patient biopsies and human CC cell lines. Using microarray expression profiling of CC cells with overexpressed or down-regulated YAP, we show that YAP regulates genes involved in proliferation, apoptosis, and angiogenesis. YAP activity promotes CC growth in vitro and in vivo by functionally interacting with TEAD transcription factors (TEADs). YAP activity together with TEADs prevents apoptosis induced by cytotoxic drugs, whereas YAP knockdown sensitizes CC cells to drug-induced apoptosis. We further show that the proangiogenic microfibrillar-associated protein 5 (MFAP5) is a direct transcriptional target of YAP/TEAD in CC cells and that secreted MFAP5 promotes tube formation of human microvascular endothelial cells. High YAP activity in human CC xenografts and clinical samples correlates with increased MFAP5 expression and CD31(+) vasculature. CONCLUSIONS: These findings establish YAP as a key regulator of proliferation and antiapoptotic mechanisms in CC and provide first evidence that YAP promotes angiogenesis by regulating the expression of secreted proangiogenic proteins.


Subject(s)
Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic , Cholangiocarcinoma/pathology , DNA-Binding Proteins/physiology , Drug Resistance, Neoplasm , Neovascularization, Pathologic/etiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Animals , Apoptosis , Bile Duct Neoplasms/blood supply , Bile Duct Neoplasms/drug therapy , Cell Cycle Proteins , Cell Proliferation , Cholangiocarcinoma/blood supply , Cholangiocarcinoma/drug therapy , Contractile Proteins/genetics , Female , Gene Expression Regulation, Neoplastic , Glycoproteins/genetics , Humans , Intercellular Signaling Peptides and Proteins , Mice , Oncogenes , TEA Domain Transcription Factors
9.
J Biol Chem ; 290(24): 14963-78, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25882841

ABSTRACT

Target of rapamycin is a Ser/Thr kinase that operates in two conserved multiprotein complexes, TORC1 and TORC2. Unlike TORC1, TORC2 is insensitive to rapamycin, and its functional characterization is less advanced. Previous genetic studies demonstrated that TORC2 depletion leads to loss of actin polarization and loss of endocytosis. To determine how TORC2 regulates these readouts, we engineered a yeast strain in which TORC2 can be specifically and acutely inhibited by the imidazoquinoline NVP-BHS345. Kinetic analyses following inhibition of TORC2, supported with quantitative phosphoproteomics, revealed that TORC2 regulates these readouts via distinct pathways as follows: rapidly through direct protein phosphorylation cascades and slowly through indirect changes in the tensile properties of the plasma membrane. The rapid signaling events are mediated in large part through the phospholipid flippase kinases Fpk1 and Fpk2, whereas the slow signaling pathway involves increased plasma membrane tension resulting from a gradual depletion of sphingolipids. Additional hits in our phosphoproteomic screens highlight the intricate control TORC2 exerts over diverse aspects of eukaryote cell physiology.


Subject(s)
Actins/metabolism , Endocytosis , Multiprotein Complexes/physiology , Saccharomyces cerevisiae/metabolism , TOR Serine-Threonine Kinases/physiology , Fungal Proteins/metabolism , Mechanistic Target of Rapamycin Complex 2 , Phosphorylation , Principal Component Analysis , Protein Kinases/metabolism , Proteomics , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction
10.
J Biomol Screen ; 19(5): 628-39, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24598104

ABSTRACT

Advances in instrumentation now allow the development of screening assays that are capable of monitoring multiple readouts such as transcript or protein levels, or even multiple parameters derived from images. Such advances in assay technologies highlight the complex nature of biology and disease. Harnessing this complexity requires integration of all the different parameters that can be measured rather than just monitoring a single dimension as is commonly used. Although some of the methods used to combine multiple measurements, such as principal component analysis, are commonly used for microarray analysis, biologists are not yet using many of the tools that have been developed in other fields to address such issues. Visualization of multiparametric data sets is one of the major challenges in this field, and a depiction of the results in a manner that can be readily interpreted is essential. This article describes a number of assay systems being used to generate such data sets en masse, and the methods being applied to their visualization and analysis. We also discuss some of the challenges of applying methods developed in other fields to biology.


Subject(s)
Computational Biology/methods , High-Throughput Screening Assays/methods , Algorithms , Chemistry, Pharmaceutical/methods , Datasets as Topic , Machine Learning , Phenotype , Principal Component Analysis , Proteins/chemistry , RNA/chemistry , Software
11.
Microbiol Res ; 169(2-3): 107-20, 2014.
Article in English | MEDLINE | ID: mdl-24360837

ABSTRACT

Due to evolutionary conservation of biology, experimental knowledge captured from genetic studies in eukaryotic model organisms provides insight into human cellular pathways and ultimately physiology. Yeast chemogenomic profiling is a powerful approach for annotating cellular responses to small molecules. Using an optimized platform, we provide the relative sensitivities of the heterozygous and homozygous deletion collections for nearly 1800 biologically active compounds. The data quality enables unique insights into pathways that are sensitive and resistant to a given perturbation, as demonstrated with both known and novel compounds. We present examples of novel compounds that inhibit the therapeutically relevant fatty acid synthase and desaturase (Fas1p and Ole1p), and demonstrate how the individual profiles facilitate hypothesis-driven experiments to delineate compound mechanism of action. Importantly, the scale and diversity of tested compounds yields a dataset where the number of modulated pathways approaches saturation. This resource can be used to map novel biological connections, and also identify functions for unannotated genes. We validated hypotheses generated by global two-way hierarchical clustering of profiles for (i) novel compounds with a similar mechanism of action acting upon microtubules or vacuolar ATPases, and (ii) an un-annotated ORF, YIL060w, that plays a role in respiration in the mitochondria. Finally, we identify and characterize background mutations in the widely used yeast deletion collection which should improve the interpretation of past and future screens throughout the community. This comprehensive resource of cellular responses enables the expansion of our understanding of eukaryotic pathway biology.


Subject(s)
Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Antifungal Agents/pharmacology , Biosynthetic Pathways , Drug Resistance, Fungal , Gene Expression Regulation, Fungal , High-Throughput Screening Assays , Molecular Sequence Data , Phylogeny , Saccharomyces cerevisiae/classification , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/metabolism
12.
Nat Biotechnol ; 29(3): 255-65, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21258344

ABSTRACT

The development of selective histone deacetylase (HDAC) inhibitors with anti-cancer and anti-inflammatory properties remains challenging in large part owing to the difficulty of probing the interaction of small molecules with megadalton protein complexes. A combination of affinity capture and quantitative mass spectrometry revealed the selectivity with which 16 HDAC inhibitors target multiple HDAC complexes scaffolded by ELM-SANT domain subunits, including a novel mitotic deacetylase complex (MiDAC). Inhibitors clustered according to their target profiles with stronger binding of aminobenzamides to the HDAC NCoR complex than to the HDAC Sin3 complex. We identified several non-HDAC targets for hydroxamate inhibitors. HDAC inhibitors with distinct profiles have correspondingly different effects on downstream targets. We also identified the anti-inflammatory drug bufexamac as a class IIb (HDAC6, HDAC10) HDAC inhibitor. Our approach enables the discovery of novel targets and inhibitors and suggests that the selectivity of HDAC inhibitors should be evaluated in the context of HDAC complexes and not purified catalytic subunits.


Subject(s)
Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Mass Spectrometry/methods , Peptide Mapping/methods , Protein Interaction Mapping/methods , Proteomics/methods
13.
Nat Biotechnol ; 25(9): 1035-44, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17721511

ABSTRACT

We describe a chemical proteomics approach to profile the interaction of small molecules with hundreds of endogenously expressed protein kinases and purine-binding proteins. This subproteome is captured by immobilized nonselective kinase inhibitors (kinobeads), and the bound proteins are quantified in parallel by mass spectrometry using isobaric tags for relative and absolute quantification (iTRAQ). By measuring the competition with the affinity matrix, we assess the binding of drugs to their targets in cell lysates and in cells. By mapping drug-induced changes in the phosphorylation state of the captured proteome, we also analyze signaling pathways downstream of target kinases. Quantitative profiling of the drugs imatinib (Gleevec), dasatinib (Sprycel) and bosutinib in K562 cells confirms known targets including ABL and SRC family kinases and identifies the receptor tyrosine kinase DDR1 and the oxidoreductase NQO2 as novel targets of imatinib. The data suggest that our approach is a valuable tool for drug discovery.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Proteomics/methods , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Benzamides , Cell Extracts , Chromatography, Affinity , Discoidin Domain Receptor 1 , Enzymes, Immobilized/antagonists & inhibitors , HeLa Cells , Humans , Imatinib Mesylate , Inhibitory Concentration 50 , K562 Cells , Pharmaceutical Preparations , Phosphorylation/drug effects , Piperazines/pharmacology , Pyrimidines/pharmacology , Quinone Reductases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL