Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 14(1): 13069, 2024 06 06.
Article in English | MEDLINE | ID: mdl-38844820

ABSTRACT

Insertion mutations in exon 20 of the epidermal growth factor receptor gene (EGFR exon20ins) are rare, heterogeneous alterations observed in non-small cell lung cancer (NSCLC). With a few exceptions, they are associated with primary resistance to established EGFR tyrosine kinase inhibitors (TKIs). As patients carrying EGFR exon20ins may be eligible for treatment with novel therapeutics-the bispecific antibody amivantamab, the TKI mobocertinib, or potential future innovations-they need to be identified reliably in clinical practice for which quality-based routine genetic testing is crucial. Spearheaded by the German Quality Assurance Initiative Pathology two international proficiency tests were run, assessing the performance of 104 participating institutes detecting EGFR exon20ins in tissue and/or plasma samples. EGFR exon20ins were most reliably identified using next-generation sequencing (NGS). Interestingly, success rates of institutes using commercially available mutation-/allele-specific quantitative (q)PCR were below 30% for tissue samples and 0% for plasma samples. Most of these mutation-/allele-specific (q)PCR assays are not designed to detect the whole spectrum of EGFR exon20ins mutations leading to false negative results. These data suggest that NGS is a suitable method to detect EGFR exon20ins in various types of patient samples and is superior to the detection spectrum of commercially available assays.


Subject(s)
Carcinoma, Non-Small-Cell Lung , ErbB Receptors , Exons , High-Throughput Nucleotide Sequencing , Lung Neoplasms , Humans , ErbB Receptors/genetics , High-Throughput Nucleotide Sequencing/methods , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Laboratory Proficiency Testing , Antibodies, Bispecific/therapeutic use , Mutagenesis, Insertional , Protein Kinase Inhibitors/therapeutic use
2.
Blood ; 139(25): 3617-3629, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35344582

ABSTRACT

Genetic alterations in the DNA damage response (DDR) pathway are a frequent mechanism of resistance to chemoimmunotherapy (CIT) in B-cell malignancies. We have previously shown that the synergy of CIT relies on secretory crosstalk elicited by chemotherapy between the tumor cells and macrophages. Here, we show that loss of multiple different members of the DDR pathway inhibits macrophage phagocytic capacity in vitro and in vivo. Particularly, loss of TP53 led to decreased phagocytic capacity ex vivo across multiple B-cell malignancies. We demonstrate via in vivo cyclophosphamide treatment using the Eµ-TCL1 mouse model that loss of macrophage phagocytic capacity in Tp53-deleted leukemia is driven by a significant downregulation of a phagocytic transcriptomic signature using small conditional RNA sequencing. By analyzing the tumor B-cell proteome, we identified a TP53-specific upregulation of proteins associated with extracellular vesicles (EVs). We abrogated EV biogenesis in tumor B-cells via clustered regularly interspaced short palindromic repeats (CRISPR)-knockout (KO) of RAB27A and confirmed that the EVs from TP53-deleted lymphoma cells were responsible for the reduced phagocytic capacity and the in vivo CIT resistance. Furthermore, we observed that TP53 loss led to an upregulation of both PD-L1 cell surface expression and secretion of EVs by lymphoma cells. Disruption of EV bound PD-L1 by anti-PD-L1 antibodies or PD-L1 CRISPR-KO improved macrophage phagocytic capacity and in vivo therapy response. Thus, we demonstrate enhanced EV release and increased PD-L1 expression in TP53-deficient B-cell lymphomas as novel mechanisms of macrophage function alteration in CIT resistance. This study indicates the use of checkpoint inhibition in the combination treatment of B-cell malignancies with TP53 loss.


Subject(s)
B7-H1 Antigen , Extracellular Vesicles , Lymphoma, B-Cell , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Extracellular Vesicles/metabolism , Lymphoma/metabolism , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Macrophages/metabolism , Mice , Neoplasms/metabolism
3.
J Neurotrauma ; 38(16): 2311-2322, 2021 Aug 15.
Article in English | MEDLINE | ID: mdl-33514282

ABSTRACT

Immunomodulation by adipose-tissue-derived stem cells (ADSCs) is of special interest for the alleviation of damaging inflammatory responses in central nervous system injuries. The present study explored the effects of cerebrospinal fluid (CSF) from traumatic brain injury (TBI) patients on this immunomodulatory potential of ADSCs. CSF conditioning of ADSCs increased messenger RNA levels of both pro- and anti-inflammatory genes compared to controls. Exposure of phorbol-12-myristate-13-acetate-differentiated THP1 macrophages to the secretome of CSF-conditioned ADSCs downregulated both proinflammatory (cyclooxygenase-2, tumor necrosis factor alpha) and anti-inflammatory (suppressor of cytokine signaling 3, interleukin-1 receptor antagonist, and transforming growth factor beta) genes in these cells. Interleukin-10 expression was elevated in both naïve and conditioned secretomes. ADSC secretome treatment, further, induced macrophage maturation of THP1 cells and increased the percentage of CD11b+, CD14+, CD86+, and, to a lesser extent, CD206+ cells. This, moreover, enhanced the phagocytic activity of CD14+ and CD86+ cells, though independently of pre-conditioning. Secretome exposure, finally, also induced a reduction in the percentage of CD192+ adherent cells in cultures of peripheral blood mononuclear cells (PBMCs) from both healthy subjects and TBI patients. This limited efficacy (of both naïve and pre-conditioned secretomes) suggests that the effects of lymphocyte-monocyte paracrine signaling on the fate of cultured PBMCs are strongest upon adherent cell populations.


Subject(s)
Brain Injuries, Traumatic/pathology , Cerebrospinal Fluid , Culture Media, Conditioned , Mesenchymal Stem Cells/physiology , Secretome/immunology , Transplantation Conditioning , Adult , Aged , Case-Control Studies , Cell Culture Techniques , Female , Humans , Inflammation , Leukocytes, Mononuclear/physiology , Macrophages/physiology , Male , Middle Aged , Young Adult
4.
Cancers (Basel) ; 12(8)2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32824276

ABSTRACT

Targeted inhibition of Bruton's Tyrosine Kinase (BTK) with ibrutinib and other agents has become important treatment options in chronic lymphocytic leukemia, Waldenström's Macroglobulinemia, Mantle cell lymphoma, and non-GCB DLBCL. Clinical trials combining small molecule inhibitors with monoclonal antibodies have been initiated at rapid pace, with the biological understanding between their synergistic interactions lagging behind. Here, we have evaluated the synergy between BTK inhibitors and monoclonal antibody therapy via macrophage mediated antibody dependent cellular phagocytosis (ADCP). Initially, we observed increased ADCP with ibrutinib, whilst second generation BTK inhibitors failed to synergistically interact with monoclonal antibody treatment. Kinase activity profiling under BTK inhibition identified significant loss of Janus Kinase 2 (JAK2) only under ibrutinib treatment. We validated this potential off-target effect via JAK inhibition in vitro as well as with CRISPR/Cas9 JAK2-/- experiments in vivo, showing increased ADCP and prolonged survival, respectively. This data supports inhibition of the JAK-STAT (Signal Transducers and Activators of Transcription) signaling pathway in B-cell malignancies in combination with monoclonal antibody therapy to increase macrophage-mediated immune responses.

5.
N Biotechnol ; 46: 14-21, 2018 Nov 25.
Article in English | MEDLINE | ID: mdl-29870785

ABSTRACT

The study of the biology and function of B cells, or the dissection and in vitro creation of enormous recombinant antibody repertoires, requires the isolation of large numbers of pure CD19+ B cells. The StraightFrom® Leukopak CD19 MicroBead Kit was recently introduced as a fast and robust kit to isolate human CD19+ B cells. This uses paramagnetic microbeads conjugated to high-affinity anti-CD19 monoclonal antibodies to bind B cells in leukapheresis (Leukopak) samples. The overall purity of the isolated cells, together with the characterization of the different CD19+ subclasses, was assessed by flow cytometry using a recombinant (REAffinity) antibody panel, revealing that the method allowed the recovery of over 93% of CD19+ B cells without any pre-purification step. This enables the relatively straightforward purification of all the circulating CD19+ B cells in a single donor.


Subject(s)
Antigens, CD19/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Separation , Flow Cytometry , Antibodies, Monoclonal/immunology , Antigens, CD19/immunology , B-Lymphocytes/immunology , Humans
6.
Cell ; 156(3): 590-602, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24485462

ABSTRACT

Therapy-resistant microenvironments represent a major barrier toward effective elimination of disseminated malignancies. Here, we show that select microenvironments can underlie resistance to antibody-based therapy. Using a humanized model of treatment refractory B cell leukemia, we find that infiltration of leukemia cells into the bone marrow rewires the tumor microenvironment to inhibit engulfment of antibody-targeted tumor cells. Resistance to macrophage-mediated killing can be overcome by combination regimens involving therapeutic antibodies and chemotherapy. Specifically, the nitrogen mustard cyclophosphamide induces an acute secretory activating phenotype (ASAP), releasing CCL4, IL8, VEGF, and TNFα from treated tumor cells. These factors induce macrophage infiltration and phagocytic activity in the bone marrow. Thus, the acute induction of stress-related cytokines can effectively target cancer cells for removal by the innate immune system. This synergistic chemoimmunotherapeutic regimen represents a potent strategy for using conventional anticancer agents to alter the tumor microenvironment and promote the efficacy of targeted therapeutics.


Subject(s)
Disease Models, Animal , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Tumor Microenvironment , Animals , Cyclophosphamide/therapeutic use , Cytokines/immunology , Drug Resistance, Neoplasm , Heterografts , Humans , Immunity, Innate , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Macrophages/immunology , Mice , Neoplasm Transplantation
7.
PLoS One ; 7(9): e45466, 2012.
Article in English | MEDLINE | ID: mdl-23029029

ABSTRACT

Macrophages are dynamic cells integrating signals from their microenvironment to develop specific functional responses. Although, microarray-based transcriptional profiling has established transcriptional reprogramming as an important mechanism for signal integration and cell function of macrophages, current knowledge on transcriptional regulation of human macrophages is far from complete. To discover novel marker genes, an area of great need particularly in human macrophage biology but also to generate a much more thorough transcriptome of human M1- and M1-like macrophages, we performed RNA sequencing (RNA-seq) of human macrophages. Using this approach we can now provide a high-resolution transcriptome profile of human macrophages under classical (M1-like) and alternative (M2-like) polarization conditions and demonstrate a dynamic range exceeding observations obtained by previous technologies, resulting in a more comprehensive understanding of the transcriptome of human macrophages. Using this approach, we identify important gene clusters so far not appreciated by standard microarray techniques. In addition, we were able to detect differential promoter usage, alternative transcription start sites, and different coding sequences for 57 gene loci in human macrophages. Moreover, this approach led to the identification of novel M1-associated (CD120b, TLR2, SLAMF7) as well as M2-associated (CD1a, CD1b, CD93, CD226) cell surface markers. Taken together, these data support that high-resolution transcriptome profiling of human macrophages by RNA-seq leads to a better understanding of macrophage function and will form the basis for a better characterization of macrophages in human health and disease.


Subject(s)
Gene Expression Profiling , Macrophages/metabolism , Transcriptome , Alternative Splicing , Cluster Analysis , Exome , Gene Expression Regulation , Gene Regulatory Networks , Humans , Immunophenotyping , Leukocytes, Mononuclear/metabolism , Lipopolysaccharide Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...