GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
Material
Language
Subjects(RVK)
  • 1
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 4303-4303
    Abstract: Introduction: Myelodysplastic syndrome (MDS) is a heterogeneous group of bone marrow disorders with a yearly incidence of approximately 13,000 in the United States. It has been observed that both genetic mutations within stem and progenitor cells and a disordered immune microenvironment are present early in MDS. Abnormal levels of inflammatory cytokines as well increased numbers of suppressive cell types, such as regulatory T cells and myeloid derived suppressor cells (MDSC) have been noted in MDS bone marrow. MDSC are recently discovered subset of myeloid cells with specific immune regulatory functions, such as T cells suppression, seen in pathological conditions, such as cancer. Recent data suggest MDSC may play a critical role in MDS pathogenesis, and that S100A9, a danger-associated molecular pattern (DAMP) produced by some myeloid cells, including neutrophils, monocytes and MDSC, is a key signal for bone marrow immune dysregulation. Here, we report a systems immunology approach to cell type discovery within MDS bone marrow using high dimensional mass cytometry. Methods: Bone marrow aspirate samples with informed consent from MDS (n=19) and AML (n=4) patients were collected and cryopreserved following red blood cell lysis for storage by the Vanderbilt Hematology Tissue Repository, a tissue repository approved by the local Institutional Review Board (IRB). Samples were acquired for the study and stained with a 35-marker panel of metal tagged mass cytometry antibodies and analyzed with a mass cytometer (CyTOF). Cellular populations were then characterized using biaxial gating as well as viSNE, SPADE and hierarchical clustering as has been previously reported (Diggins et al. Methods 2015, Ferrell et al. PLoS One, 2016). Results: Unsupervised viSNE analysis of 35-markers per cell revealed distinct cellular subsets within each sample. Interestingly, one of the strongest marker signals was expression of S100A9, which was seen in multiple cells types including phenotypic MDSC. Further analysis revealed that as a percentage of bone marrow cells, S100A9 expression was significantly more common in RCMD vs. RAEB and AML (30.0% (n=10) vs. 10.9% (n=9) and 2.4% (n=4), respectively, p 〈 0.05 for each comparison) (Figure 1A). Additionally, three paired RCMD/AML samples were available for analysis. Within these patients, the percentage of S100A9+ cells dropped from a mean of 41.7% in RCMD to a mean of 1.84% in AML bone marrow (Figure 1B & C). Conclusion: S100A9 is both a distinguishing feature of RCMD and of disease progression within MDS. Because of its important role inflammation and cellular recruitment, S100A9 may correlate with bone marrow cellular inflammation and could represent a viable target in treatment of the disordered immune microenvironment present in MDS, especially RCMD. Disclosures Savona: Celgene: Membership on an entity's Board of Directors or advisory committees; Sunesis: Research Funding; Incyte: Membership on an entity's Board of Directors or advisory committees, Research Funding; Ariad: Membership on an entity's Board of Directors or advisory committees; Amgen Inc.: Membership on an entity's Board of Directors or advisory committees; TG Therapeutics: Research Funding; Takeda: Research Funding; Gilead: Membership on an entity's Board of Directors or advisory committees. Irish:Incyte: Research Funding; Janssen: Research Funding; Cytobank, Inc.: Equity Ownership, Membership on an entity's Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 3686-3686
    Abstract: Background: Acute myeloid leukemia (AML) is the most common acute leukemia in adults and has a poor outcome with limited treatment options in patients with relapsed or resistant disease. Therapy resistance in AML is likely related to the inadequacy of therapy within leukemia cell subsets, including leukemia stem cells (LSCs). The p21-activated kinase (PAK) family of proteins was shown to be overexpressed in cancer cells and to play a key role in proliferation, survival, and maintenance of cellular structure. The series of orally bioavailable PAK4 allosteric modulators (PAM) have previously been shown to have activity in hematological cancer cell lines, including those derived from acute myeloid leukemia (AML) (Senapedis et al. Blood124, 2208-2208). Understanding how therapies target cellular subsets within primary patient samples could aid drug development by revealing any subset specific drug effects. In this project, we studied the effects of p21-activated kinase 4 (PAK4) modulation in AML samples. PAK4 modulation has been shown to have significant effects on many intracellular signaling pathways, including PI3K/AKT, MAPK/ERK and WNT/β-catenin pathways (Senapedis et al. Blood124, 2208-2208). It is unknown whether PAMs will have similar activity in primary leukemia cells. Likewise, it is currently unclear to what extent PAMs will differentially impact primary cell subsets including leukemia stem cells and non-malignant cell subsets that may be critical to recovery of bone marrow functions. We have previously shown that the single cell biology platform of flow cytometry is well-suited for dissecting clinically relevant signaling network mechanisms in primary human AML (Irish et al. Cell, 118(2):217-28). Methods: Flow cytometry was used to dissect the impact of an orally bioavailable PAM in AML cell lines and primary patient tissue. Cell lines chosen for this study included NRAS mutant KG-1 and Kasumi-1, which carry t(8;21) and express the AML1:ETO fusion protein. Primary AML biopsies were acquired from bone marrow or blood prior to any treatment and patients were identified and consented for this study according to a local Institutional Review Board-approved protocol. AML tissue samples were viably cryopreserved and then assayed ex vivo. Established protocols were used for phospho-specific flow cytometry, fluorescent cell barcoding, and data analysis in Cytobank (Irish et al. Cell, 118(2):217-28, Doxie and Irish, Curr Top Microbiol Immunol. 377:1-21). Results: Differential effects of PAK4 inhibition were observed between cell lines and among cell subsets from AML patient bone marrow. In leukemia cell lines and patient samples, p-ERK and p-S6 showed marked inhibition via PAM, though degree of inhibition varied. In AML patient samples, PAMs blocked signaling responses in p-ERK specifically in AML blasts, but spared normal CD45hi mononuclear cells (0.88 vs. 0.29-fold reduction (arcsinh scale) in p-ERK at 10 nM). Within the AML blast population, CD34+ CD38- and CD34+ CD38+ AML subsets showed similar PAM dose response via p-ERK. Conclusions: Single cell analysis effectively distinguishes effects of PAK4 inhibition via a series of allosteric modulators of PAK4 (PAMs) on leukemia and non-leukemia subsets in the same sample. PAM reduced immediate p-ERK and p-S6 levels in primary leukemia and cell lines. Notably, inhibition in various subsets within human AML was successfully measured by phospho-flow cytometry. Signaling changes in p-ERK were minimal within non-leukemic mature CD45+ mononuclear cells found in primary patient biopsies. Analysis of CD34+ CD38- cells indicates that PAMs could have activity within leukemia stem cells, and, at least, effect the AML progenitors. These findings support further investigation into the mechanism of action and treatment potential of PAMs in AML. Disclosures Senapedis: Karyopharm Therapeutics, Inc.: Employment, Patents & Royalties. Baloglu:Karyopharm Therapeutics Inc.: Employment, Equity Ownership. Landesman:Karyopharm: Employment. Irish:Novartis: Honoraria; Cytobank, Inc.: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Karyopharm: Research Funding; InCyte: Research Funding. Savona:Gilead: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Incyte: Membership on an entity's Board of Directors or advisory committees, Research Funding; Karyopharm: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 148-148
    Abstract: Abstract 148 Background: Successful therapeutic targeting of genetic lesions in cancer generally requires that such lesions be clonally dominant and uniformly represented. The commonest indolent non-Hodgkin's lymphoma (NHL) subtype, Follicular lymphoma (FL), is currently incurable with conventional regimes and is therefore an attractive candidate for targeted therapies. Our prior studies[1] and studies by others[2] ,[3] have shown that clinically significant variation in patient outcomes can be captured by differences in expression of CD20 both between tumor B-cells and between patients with NHL treated with or without rituximab. We hypothesized that a hierarchy of somatic mutations might exist in FL, and could impact the opportunity for future mutation-directed therapies in this disease. Methods: Here, we examined clonal diversity in human Follicular Lymphoma (FL) using tumor subpopulations at diagnosis and at disease progression, through a genetic analysis of coding genomes and gene expression. We profiled whole-exomes and transcriptomes of 26 tumor-derived subpopulations sorted based on expression of CD20 in 10 lymphomas derived from 8 patients, including pairs at diagnosis and first disease relapse (mean exome coverage per patient 〉 200x). We used phylogenies derived from ongoing somatic hypermutation of cloned immunoglobulin VH genes at diagnosis and relapse as a framework for comparing mutational hierarchies across the coding genome. We employed qPCR of invariant BCL2 and VDJ recombination to quantitate tumor purity in subpopulations. Results: We identified 882 somatic nucleotide variants (SNVs) encoding missense and nonsense mutations in 572 unique genes, with an average of 103 mutations/case, with 95% of mutations seen in only 1 case. Mutated genes were significantly enriched for those involved in chromosome and chromatin organization (FDR=4.07×10-4 and FDR=6.40×10-4, respectively). We identified mutations in many of the genes previously implicated in lymphomas, including MLL2 and CREBBP, and in several genes not previously implicated in lymphomas including IKZF2, CD40, CALR, NBPF14, ROS1 and ERBB2. We observed 11–343 nonsynonymous coding mutations per tumor, with a striking tendency for most of these to be subclonal based on their low allelic variant frequencies (Figure 1). Many of these mutations were differentially distributed between tumor subpopulations which were evident both at diagnosis and at relapse. In addition, mutations in these populations were associated with distinct gene expression profiles. Surprisingly, subclonal distribution of mutations occurred in several genes which are known to be highly recurrently mutated in FL and thought to be drivers of pathogenesis. With rare exception, the frequency of mutation within a given gene across 375 patients with NHL studied by high throughput sequencing (FL=24, DLBCL=158, CLL=193) correlated poorly with clonal dominance within individual FL tumors in this study. By using immunoglobulin somatic mutations and BCL2 translocations as a frame of reference and by comparing diagnosis and relapse tumor pairs, we could distinguish early versus late somatic events during tumor evolution. We observed two contrasting patterns of clonal evolution, reflecting either maintenance or loss of most mutations at relapse. These two patterns were also mirrored by maintenance or loss of DNA copy number alterations in the respective cases. Conclusions: These observations allow construction of genetic evolution models for FL. This framework provides important insight into lymphomagenesis and is a key step in prioritization of candidates for gene mutation-directed therapies. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 4572-4572
    Abstract: Introduction: Allogeneic stem cell transplant (SCT) fundamentally alters the immune milieu of an individual and provides a curative treatment for patients with blood cancer. However, a common side effect of SCT is the development of chronic graft versus host disease (cGVHD). Given the growth of new experimental therapeutics for GVHD, a systems immune monitoring approach based on mass cytometry analysis of peripheral blood may provide a robust and minimally invasive way to characterize the post-SCT cGVHD immune environment and systematically track cellular biomarkers of immune status, disease severity, or ongoing treatment responses (Greenplate et al., Euro J Cancer 2016, Kordasti et al., Blood 2016). It may also be possible to identify shifts in the immune milieu or cell signaling that predict treatment responses or stratify risks (Greenplate et al., Cancer Immunol Res 2016). This study tested the feasibility of using mass cytometry peripheral blood analysis in clinical research with the goals of 1) better understanding the post-SCT and cGVHD immune environment and 2) developing a knowledge base for monitoring treatment responses. Methods: Peripheral blood mononuclear cells (PBMC) were collected with informed consent from cGVHD patients or healthy donors with approval of the local Institutional Review Board (IRB) and in accordance with the Declaration of Helsinki. A total of 88 viably cryopreserved PBMC samples were analyzed from 11 individual patients undergoing extracorporeal photopheresis (ECP) therapy. For each patient, PBMC from 4 clinical timepoints representing pre-ECP and 2, 4, and 6 months post-ECP were characterized with 2 systems immune monitoring antibody panels emphasizing B cells and T cells (Table 1). Staining, data collection using a CyTOF mass cytometer, bead based normalization, and analysis with Cytobank software followed established protocols (Leelatian et al., Methods Mol Bio 2016, Polikowsky et al., J Immunol 2015). Next, cell subsets were algorithmically revealed and characterized by computational tools (Diggins et al., Methods 2015) and patterns in the abundance and phenotype of T, B, NK, and myeloid cell subsets systematically compared to patient clinical features. Results: Single cell tools revealed profound, abnormal shifts in the peripheral immune environment in individual patients that contrasted with relatively stable immunophenotypes observed in healthy individuals and prior studies. Expected immune changes, such a profound loss of B cells post-treatment with Rituximab, were readily detected by computational tools and expert review. Expression of signaling, activation, and subset identity markers in B and T cell subsets displayed striking variation between patients and was effectively quantified on at least 5 major cell subsets and numerous minor cell subsets (Figure 1). These data highlighted how computational tools paired with high-dimensional mass cytometry can systematically characterize key protein features and cell subsets in complex and heterogeneous diseases including cancer and cGVHD and provided a rationale for ongoing association analyses between cellular subsets and clinical outcomes. Conclusion: Systems immune monitoring successfully revealed shifts in cell abundance and immunophenotype pre- and post-ECP for patients with cGVHD. These shifts were detected by computational tools and revealed alterations to the immune system following treatment. This study indicates that single cell quantitative analysis is feasible as a clinical research tool to characterize immunotherapy treatment responses, and reveal cellular biomarkers. Integration of such tools in prospective clinical trials is essential to establish and once validated, may augment physician decision making. Disclosures Polikowsky: Cytobank: Employment. Greenplate:Cytobank: Consultancy. Jagasia:Therakos: Consultancy. Irish:Cytobank, Inc.: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Janssen: Research Funding; Incyte: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 136, No. Supplement 1 ( 2020-11-5), p. 15-16
    Abstract: Background: Cytomegalovirus (CMV) serostatus is associated with transplant related mortality, relapse and survival following allogeneic stem cell transplant (SCT). Prior studies have demonstrated that CMV seronegative recipients (R-) receiving a CMV seropositive graft (D+) experience inferior outcomes compared to the R-/D- combination, an observation that is independent of viral reactivation and due to increased bacterial and fungal sepsis (Nichols WG et al, J. Infect. Dis. 2002; 185:273-82). We therefore investigated the hypothesis that CMV exposure, including that by the donor only prior to transplant, promotes endogenous and long-term immunodeficiency after SCT. Methods: A total of 108 patient peripheral blood samples were obtained from the Chronic Graft-vs.-Disease (GVHD) Consortium, which includes both patients with and without chronic GVHD. Seventy-three baseline samples were taken at an average of 458 days (range 63 to 1,965) after SCT, and 35 paired blood samples were taken at an average of 94 days follow-up. Transplant parameters, CMV serostatus, and CMV viral titers after SCT were collected. We performed multiparameter flow cytometry to assess innate and adaptive immunity after SCT in the context of CMV, including transcription factor and cytokine expression, together with TCR/BCR sequencing to assess immune diversity. Results: We demonstrate that CMV exposure (including solely in the donor prior to transplant or in the recipient after transplant) is strongly associated with long-lasting expansion of a terminally differentiated MHC class II-restricted donor memory T cell subset identifiable by the surface markers CD4+/CD57+/CD27-(Figure 1). These cells are CCR7neg/CD45RO+/CD45RA+/-, express T-bet, eomesodermin (EOMES), granzyme B, and secret high amounts of Th1 cytokines IFNγ and TNF (Figure 1). This T cell subset represents an average of 2.8% for D-/R- transplants and & gt;10% (and up to 70%) of total CD4+ T cells in all other serostatus combinations irrespective of the presence or absence of measurable CMV reactivation after SCT (p & lt;0.05 across all pairwise comparisons with D-/R- subset, Figure 2). Furthermore, this phenotype persists in an expanded fashion for years following SCT (samples were taken up to 5 years post-SCT). Follow-up samples from the same patient taken up to 218 days after the initial sample confirm phenotypic stability over time (r=0.90, p & lt;0.0001, Figure 3). Detectable CMV replication after SCT further augmented this memory CD4+ T cell subset, and antiviral treatment for CMV did not limit its expansion. We demonstrate that these differentiated memory CD4+ T cells are associated with a significant loss of T cell receptor diversity (r=-0.60, p=0.004) and reduced proportions of major histocompatibility class (MHC) II expressing classical monocytes (r=-0.43, p=0.0002) and plasmacytoid dendritic cells (r=-0.31, p=0.0085) in peripheral blood. Conclusion: These data describe a temporally stable, expanded MHC class II-restricted memory T cell population with high cytotoxic potential and putative mechanisms by which CMV exposure modifies immune function in the peri-transplant period, namely the limitation of TCR diversity and antigen presentation. This cell population is expanded even in the absence of detectable CMV reactivation post SCT and can be observed for several years following transplant. We thus provide a potential mechanism for the immunodeficiency and broad increase in infection mortality invoked by CMV exposure. In particular, these findings may provide a rationale for supporting the selection of CMV- donors for R- SCT recipients. Disclosures Jagasia: Ocugen: Other; Mallinckrodt: Research Funding; Janssen: Research Funding. Dahlman:Kadmon Holdings, Inc: Other. Lee:Syndax: Research Funding; Pfizer: Consultancy, Research Funding; AstraZeneca: Research Funding; Amgen: Research Funding; Kadmon: Research Funding; Takeda: Research Funding; Novartis: Research Funding; Incyte: Consultancy, Research Funding. Hill:Generon: Consultancy; Roche: Research Funding; Compass Pharmaceuticals: Research Funding; CSL: Research Funding; Implicit Bioscience: Research Funding; Pharmacyclics: Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2020
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: PLOS ONE, Public Library of Science (PLoS), Vol. 11, No. 4 ( 2016-4-13), p. e0153207-
    Type of Medium: Online Resource
    ISSN: 1932-6203
    Language: English
    Publisher: Public Library of Science (PLoS)
    Publication Date: 2016
    detail.hit.zdb_id: 2267670-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cytometry Part B: Clinical Cytometry, Wiley, Vol. 92, No. 1 ( 2017-01), p. 79-87
    Abstract: Follicular lymphoma (FL) is an indolent non‐Hodgkin lymphoma that has a risk of transformation to more aggressive lymphoma. Relatively little is known about the nonmalignant B‐cell and T‐cell subset composition within the tumor microenvironment and whether altered phenotypes are associated with patterns of lymphoma B‐cell heterogeneity. Methods Two mass cytometry (CyTOF) panels were designed to immunophenotype B and T cells in FL tumors. Populations of malignant B cells, nonmalignant B cells, and T cells from each FL tumor were identified and their phenotypes compared to B and T cells from healthy human tonsillar tissue. Results Diversity in cellular phenotype between tumors was greater for the malignant B cells than for nonmalignant B or T cells. The malignant B‐cell population bore little phenotypic similarity to any healthy B‐cell subset, and unexpectedly clustered closer to naïve B‐cell populations than GC B‐cell populations. Among the nonmalignant B cells within FL tumors, a significant lack of GC and plasmablast B cells was observed relative to tonsil controls. In contrast, nonmalignant T cells in FL tumors were present at levels similar to their cognate tonsillar T‐cell subsets. Conclusion Mass cytometry revealed that diverse HLA‐DR expression on FL cells within individual tumors contributed greatly to tumor heterogeneity. Both malignant and nonmalignant B cells in the tumor bore little phenotypic resemblance to healthy GC B cells despite the presence of T follicular helper cells in the tumor. These findings suggest that ongoing signaling interactions between malignant B cells and intra‐tumor T cells shape the tumor microenvironment. © 2016 International Clinical Cytometry Society
    Type of Medium: Online Resource
    ISSN: 1552-4949 , 1552-4957
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2017
    detail.hit.zdb_id: 2180651-2
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 144-144
    Abstract: Abstract 144 Introduction: Mantle cell lymphoma (MCL) is an aggressive non-Hodgkin's lymphoma. Although newer treatment regimens including intensive chemo-immunotherapy followed by stem cell transplant have improved survival to a median approaching 6 years, MCL is still incurable in many cases. The use of flow cytometry to investigate stimulation induced signaling at the single cell level represents an opportunity to obtain each patient's signaling profile as well as to discover tumor cell heterogeneity and identify signaling events potentially responsible for poor outcomes. We recently used this method to characterize signaling in subpopulations of tumor samples from patients with follicular lymphoma (FL). In FL, we identified a lymphoma negative prognostic (LNP) cell subset with impaired B cell antigen receptor (BCR) signaling, the prevalence of which correlated with adverse clinical outcome (Irish et al., PNAS 2010). In the present study we used the same approach to identify signaling responses with large variation within MCL patient samples, and tested whether these signaling events might predict patients' clinical outcomes. By understanding the biology based on lymphoma signaling profile, we might identify prognostic significant factors such as the LNP subset observed in FL. Method: Single cell flow cytometry measurements of signaling were acquired for samples of MCL (n=25). Of these, 21 MCL specimens were obtained prior to any treatment. Median age was 61 years, and follow up time ranged from ½ until 15 years. Treatment regimens varied. Samples from diffuse large B cell lymphoma (DLBCL, n=12), FL (n=14), chronic lymphocytic leukemia (CLL, n=14) and tonsils from healthy donors (n=4) were also included for comparison. Phosphorylation of 14 signaling proteins was measured under 12 different stimulation conditions in every cell within the patient specimens, including lymphoma B cells and in tumor-infiltrating T cells. Stimulation conditions included BCR crosslinking, CD40 ligand, CpG oligodeoxynucleotides (CpG ODN) and several cytokines (IL-4, IL-10, IL-21, IFN-γ). Result: The magnitude of most cytokine induced signaling responses in lymphoma cells from MCL patients was higher than in lymphoma cells from FL. This included IL-10 induced phosphorylation of STAT3 (p 〈 0.0001) and IL-4 induced phosphorylation of STAT6 (p 〈 0.0001). In contrast, IL-21 induced phosphorylation of STAT3 in lymphoma cells was similar in FL and MCL patients. Furthermore, MCL tumor cells responded better to BCR crosslinking at 9 measured downstream signaling proteins, including SYK, Src family kinases (SFK) and PLC-γ, than tumor cells from FL and CLL patients. Although signaling responses overall were higher in MCL, there was a large variation in the lymphoma cells' signaling capacity within each lymphoma histology. To identify possible signaling events that correlated with clinical outcome in MCL, we first identified the signaling events with the largest variance. This included phosphorylation of SYK and PLC-γ by BCR crosslinking, phosphorylation of NF-κB (p65) induced by CpG ODN, CD40L, or by PMA and ionomycin, and phosphorylation of STAT3 induced by IL-10 and IL-21. Of these seven signaling events, only CpG ODN induced p-p65 significantly stratified overall survival when MCL patients were split into two even groups based on higher and lower signaling response, compared to healthy B cells from tonsils. Hence, patients whose MCL cells displayed high CpG ODN induced NF-κB (p65) signaling had an adverse prognosis (p 〈 0.008, HR=5.1). In contrast with our observations of FL, BCR signaling did not stratify the outcome of MCL patients. Conclusion: Potentiated NF-kB p65 signaling in response to CpG ODN was associated with poor prognosis in MCL. The tumor cells capacity to respond to CpG induced signaling likely represents a biological advantage for the tumor cells in this lymphoma entity. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Society of Hematology ; 2016
    In:  Blood Vol. 128, No. 22 ( 2016-12-02), p. 4296-4296
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 4296-4296
    Abstract: Background MDS is characterized by ineffective haematopoiesis and a propensity to leukaemia transformation, with increasing evidence linking immune exhaustion to disease progression. Immune checkpoints are known to be upregulated on T cells in cancer, and inhibitors of CTLA-4 and PD1/PLD1 axis have demonstrated efficacy with likely clinical benefit in MDS. We have previously shown profound changes in both the number and function of components of the adaptive immune system, particularly Tregs, in MDS (Kordasti, Blood 2007). In order to characterise the immune signature in a wider range of T cell subsets simultaneously, with particular emphasis on cells likely to be affected by checkpoint inhibitor therapy (CPI), we analyse CTLA-4 and PD1 expression in MDS by cytometry by time-of-flight (CyTOF). Additionally, we aim to explore whether these differences are accentuated by the absence or presence of somatic mutations, or in morphologically more advanced disease. Materials and Methods MDS patients (n=56) and age-matched healthy donors (HD, n=6) were stained with two panels of 35 and 34 antibodies for unstimulated and PMA/Ionomycin-stimulated PBMCs, respectively. Samples were run on CyTOF and data analysed using visual stochastic neighbour embedding (viSNE, Cytobank) to generate t-distributed SNE scores by unsupervised multi-dimensional reduction of T cells. Spanning-tree progression analysis of density-normalized events (SPADE), was performed and T cell subsets identified from heat maps based on typical phenotypic markers (Regulatory, Naive, Memory, Effector Memory (EM) Central Memory (CM), Effector, Terminal Effector (TE)). T cells were also clustered based on cytokine secretion (IFN-ϒ, TNF-α, IL-17, IL-2 and IL-10). Somatic mutation analysis was performed on 48 of the MDS patients using our established targeted panel of 24 genes known to be mutated in MDS, for subgroup analysis (Mohamedali, Leukaemia 2015) Results and Discussion Demographics and subgroups are outlined in figure 1. The number of Tregs was significantly higher in RAEB than non-RAEB MDS (9.6% of total CD4+ cells vs 7.5% p=0.02) and in RAEB versus HD (9.6% vs 5.8% p=0.01). There was a significantly higher proportion of Tregs in MDS patients with somatic mutations compared to those without (8.7% vs 7.06% p 〈 0.05) and HD (8.7% vs 5.9% p 〈 0.05), confirming our previous findings. Amongst two subpopulations of Tregs previously identified (Kordasti, Blood 2016) there was no difference between HD and MDS in terms of their number. However, Tregs A and B have an increased PD1 expression in MDS vs HD (p=0.03 & p=0.003). In addition, CD4+EM and CD4+Memory, CD4+/CD8+ Na•ve T cells and TNF-α secreting cells all expressed more PD1 when compared to HD (p 〈 0.05), see figure 2. However, there were no differences in T cell PD1 expression when comparing low and high risk MDS patients, suggesting immune exhaustion to tumour antigens is a common founder event in MDS. By contrast, CTLA-4 expression was not significantly different in T cell subsets between MDS and HD. Sub-groups based on specific somatic mutations did reveal that CD4+ Memory, CD4+TE and CD8+CM subsets in SF3B1-mutated MDS cases had less CTLA-4 expression compared to MDS cases with other mutations (p 〈 0.05). Furthermore, SF3B1-mutant cases also had less CTLA-4 expression when compared to MDS cases without any detected mutations in CD4+ Memory, CD4+EM and CD8+CM subsets (p 〈 0.05). Taken together this work suggests a possible 3 hit-model for disease progression in MDS. The establishment of MDS is accompanied by increased expression of PD1 most likely in response to increased PDL1 on tumour cells, which argues for early introduction of PD1 inhibitor in the disease course. Increased CTLA-4 expression is likely a later step, dependent on the acquisition of specific mutations by the malignant clone, that further alters the relationship between tumour and immune surveillance. Finally, in disease progression through from non-RAEB to RAEB/AML there is an expansion of suppressive Tregs facilitating tumour outgrowth. Furthermore, pro-inflammatory IL17 secreting T helper cells (Th17) have been shown to be increased in low risk MDS and are known to be associated with autoimmune disease. The trend towards increased PD1 expression in Th17 cells in a group of low risk patients harbouring SF3B1 mutations (p=0.09) is suggestive that acquisition of increased PD1 expression is a key branch point in disease outcome. Disclosures Irish: Incyte: Research Funding; Janssen: Research Funding; Cytobank, Inc.: Equity Ownership, Membership on an entity's Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2014
    In:  Nature Immunology Vol. 15, No. 12 ( 2014-12), p. 1095-1097
    In: Nature Immunology, Springer Science and Business Media LLC, Vol. 15, No. 12 ( 2014-12), p. 1095-1097
    Type of Medium: Online Resource
    ISSN: 1529-2908 , 1529-2916
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2014
    detail.hit.zdb_id: 2026412-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...