Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 101-103
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 799-800
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood Advances, American Society of Hematology, Vol. 2, No. 13 ( 2018-07-10), p. 1572-1579
    Abstract: TL in LSCs is significantly shortened at diagnosis of CML and correlates with LSC burden. TL in nonleukemic myeloid cells in deep molecular remission is unaffected by long-term TKI treatment.
    Type of Medium: Online Resource
    ISSN: 2473-9529 , 2473-9537
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 2876449-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Nature Communications, Springer Science and Business Media LLC, Vol. 9, No. 1 ( 2018-04-19)
    Abstract: Aggressive natural killer-cell (NK-cell) leukemia (ANKL) is an extremely aggressive malignancy with dismal prognosis and lack of targeted therapies. Here, we elucidate the molecular pathogenesis of ANKL using a combination of genomic and drug sensitivity profiling. We study 14 ANKL patients using whole-exome sequencing (WES) and identify mutations in STAT3 (21%) and RAS-MAPK pathway genes (21%) as well as in DDX3X (29%) and epigenetic modifiers (50%). Additional alterations include JAK-STAT copy gains and tyrosine phosphatase mutations, which we show recurrent also in extranodal NK/T-cell lymphoma, nasal type (NKTCL) through integration of public genomic data. Drug sensitivity profiling further demonstrates the role of the JAK-STAT pathway in the pathogenesis of NK-cell malignancies, identifying NK cells to be highly sensitive to JAK and BCL2 inhibition compared to other hematopoietic cell lineages. Our results provide insight into ANKL genetics and a framework for application of targeted therapies in NK-cell malignancies.
    Type of Medium: Online Resource
    ISSN: 2041-1723
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2018
    detail.hit.zdb_id: 2553671-0
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 2921-2921
    Abstract: INTRODUCTION Natural killer (NK) cell malignancies are rare aggressive neoplasms that are classified by the WHO as extranodal NK/T-cell lymphoma, nasal type (NKTCL) and aggressive NK-cell leukemia (ANKL). Recently, genome and exome level studies in NKTCL have shed light on the mutational spectrum of the disease. However, somatic mutations in ANKL have not been characterized. Here, we identified somatic mutations in 14 cases of ANKL to further clarify the genetic landscape underlying malignant NK cell proliferation. We compared the discovered variants to those detected in NKTCL to understand whether the two diseases harbor common molecular alterations. Moreover, we used high-throughput drug screening and RNA sequencing on NK cell lines derived from ANKL, NKTCL and other malignant NK cell proliferations to identify therapeutically actionable drivers of malignant NK cell growth. METHODS We performed whole-exome sequencing on genomic DNA extracted from peripheral blood or bone marrow samples of 14 ANKL patients. To compare the mutational profiles in ANKL and NTKCL, we re-analyzed the published whole-exome NKTCL data from Jiang et al. (Nat Genet 2015) using our somatic variant calling pipeline. For profiling of drug responses, we used a high-throughput drug sensitivity and resistance testing (DSRT) platform comprising 461 approved and investigational oncology drugs to screen the ANKL cell lines IMC-1, KHYG-1 and NK-92, NKTCL cell lines NK-YS and SNK-6 as well as DERL-7, KAI3, NKL, YT and IL-2-stimulated NK cells from healthy donors. All drugs were tested in 384-well format in 5 concentrations over a 10,000-fold concentration range for 72 h, cell viability was measured and normalized dose response curve values were used to calculate a drug sensitivity score (DSS) for each drug. Finally, we performed amplicon sequencing of known cancer driver genes and RNA sequencing on the cell lines and healthy NK cells to identify driver mutations and integrate gene expression profiles with drug sensitivity patterns. RESULTS We identified recurrent somatic activating mutations in STAT3 in 21% (3 of 14) of ANKL patients. Other mutated genes included RAS-MAPK pathway molecules (BRAF, NRAS, KRAS), protein phosphatases regulating JAK-STAT and PI3K-AKT-mTOR pathways (PTPRT, PTPRK, INPP5D) as well as several epigenetic modifiers (TET2, ARID2, KDM2B, SETD7, SETD2) and the tumor suppressor TP53. Interestingly, we detected mutations in genes recurrently mutated in NKTCL, such as the RNA helicase DDX3X and the cell surface receptor FAS. Re-analysis of the published NKTCL data revealed a high frequency of missense mutations in receptor type and non-receptor type protein phosphatases (e.g. PTPRC, PTPRR, PTPRT, PTPN1, PTPN2, PTPN3), many of which have established roles as negative regulators of JAK-STAT signaling. These findings potentially expand the subset of NK cell tumors where the JAK-STAT pathway is somatically activated and implicate deregulated JAK-STAT signaling as a major driver in these diseases. The malignant NK cell lines used in drug sensitivity profiling frequently harbored mutations in same genes and pathways, including STAT3 (N=3), STAT5B (N=1), DDX3X (N=2), KRAS (N=1), FAS (N=2) and several epigenetic modifiers, thus validating these cell lines as relevant disease models. The drug sensitivities in NK cell lines showed a high correlation and the cell lines formed a distinct group from other lymphoid and myeloid leukemia cell lines in unsupervised hierarchical clustering, suggesting an NK-cell specific drug response pattern. The most effective targeted drugs across all NK cell lines included HDAC inhibitors, inhibitors of Aurora and Polo-like kinases, JAK inhibitors, HSP inhibitors and CDK inhibitors as well as the Bcl-2 inhibitor navitoclax. Compared to other leukemia and lymphoma cell lines, JAK inhibitors, navitoclax and methotrexate emerged as the most NK-cell specific compounds. CONCLUSIONS Our genetic data demonstrate extensive heterogeneity in the mutational spectrum of ANKL and implicate JAK-STAT and RAS-MAPK signaling as well as disruption of epigenetic modifiers in the disease pathogenesis. Integrated drug sensitivity and gene expression profiling corroborates the JAK-STAT pathway as a major therapeutically actionable driver of malignant NK cell proliferation and identifies other potential novel targeted therapy options such as Bcl-2 inhibition in NK cell malignancies. Disclosures Suzumiya: Chugai: Honoraria, Research Funding; Toyama Chemical: Research Funding; Kyowa Hakko kirin: Research Funding; Astellas: Research Funding; Eisai: Honoraria, Research Funding; Takeda: Honoraria. Ohshima:Chugai: Research Funding, Speakers Bureau; Kyowa Kirin: Research Funding, Speakers Bureau. Mustjoki:Pfizer: Honoraria, Research Funding; Ariad: Research Funding; Novartis: Honoraria, Research Funding; Bristol-Myers Squibb: Honoraria, 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 1924-1924
    Abstract: Introduction: Progression of chronic myeloid leukemia (CML) to blast crisis (BC) results from the acquisition of additional driver mutations, which still are poorly understood. In the tyrosine kinase inhibitor (TKI) era, BC-CML remains a challenging clinical entity with very poor prognosis and short survival. In addition, there is an unmet need for identification of progression-related genetic mutations and potentially targetable pathways. Patients and methods: Bone marrow samples from 16 patients with BC-CML (myeloid n=13, lymphoid n=2, unknown phenotype n=1) and 2 patients with accelerated phase (AP) were collected from the Helsinki University Hospital, Finland and National Cancer Institute, Cairo University, Egypt. In addition, skin biopsy samples were collected for germline variant controls. Whole exome sequencing (WES) (n=8) was done with Agilent or NimbleGen exome capture kits and deep-targeted sequencing with the NimbleGen (SeqCap EZ Design) comprehensive cancer gene panel (n=10) using an Illumina HiSeq instrument. The panel was comprised of 578 driver genes with documented association to common and rare cancers (gathered from Sanger and NCBI tests databases). All mutated genes identified by WES were included in the panel, as well as genes previously reported to be mutated in BC-CML. Results: We identified 55 mutations in 33 driver genes (average: 3 per patient, range 0-7). Of the identified 33 mutated genes, 27 were ranked ≥1 in the Gene Ranker Cancer scoring system (http://cbio.mskcc.org/tcga-generanker), where genes with a score of 1 have a documented association with cancer in a cancer gene database and higher scores indicate more frequent incidence of gene mutations in different cancers. Core-binding factor (CBF) aberrations (RUNX1 mutations and inv [16]) were the most recurrent variants (n=6 in 5 patients, 27.7% of the patients) followed by ABL1 mutations (n=4 in 4 patients, 22%) and BCOR mutations (n=4 in 3 patients, 16.7%). Other recurrent mutations included FLT3, IKZF1, and NOTCH1 mutations which all were found in 2 cases. Some of the discovered mutations have not been reported in BC-CML patients before, such as mutations to MTOR, PTPRJ, CD274 (PD-L1), IL21R, SETD2 and ZRSR2 genes. In silico analysis of the targeted genes showed that many of the affected genes interact with each other in different pathways and also with ABL1. The top pathways affected were associated with key biological functions: regulation of hematopoiesis (11 genes affected), leucocyte differentiation (9 genes) and transcriptional regulation (11 genes). Conclusion: The genomic landscape of advanced phases of CML (BC and AP) shows complex heterogeneity with a broad range of genes affected leading to dysregulation of multiple molecular pathways that have an impact on treatment responses and disease biology. Such complexity suggests that a personalized approach maybe the best treatment option for these patients. Disclosures Heckman: Celgene: Research Funding; Pfizer: Research Funding. Porkka:Novartis: Honoraria, Research Funding; Bristol-Myers Squibb: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding. Mustjoki:Pfizer: Honoraria, Research Funding; Ariad: Research Funding; Novartis: Honoraria, Research Funding; Bristol-Myers Squibb: Honoraria, 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Oncotarget, Impact Journals, LLC, Vol. 8, No. 36 ( 2017-09-01), p. 60123-60134
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2017
    detail.hit.zdb_id: 2560162-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Clinical Immunology, Elsevier BV, Vol. 175 ( 2017-02), p. 75-81
    Type of Medium: Online Resource
    ISSN: 1521-6616
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2017
    detail.hit.zdb_id: 1462862-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 190-190
    Abstract: Introduction: The oncoprotein Bcr-Abl has two major isoforms, depending on the breakpoint in BCR gene, p190 and p210. While p210 is the hallmark of chronic myeloid leukemia (CML), p190 occurs in the majority of Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL) patients. p190 occurs as a sole transcript in 1-2% CML patients, associated with distinct features like monocytosis and frequent additional cytogenetic abnormalities (ACA) at diagnosis. It also confers a risk of treatment failure and progression in chronic phase (CP) CML patients. However, the underlying mechanisms are largely unknown. Here we explore the characteristics of p190 and p210 in CML and ALL patients using next generation sequencing, phospho-flowcytometry and high throughput drug testing. Patients and methods: Peripheral blood mononuclear cells (PMNC) were collected at diagnosis from four CP-CML patients harboring p190 isoform from Helsinki University Hospital. Genetic alterations were identified by whole exome sequencing. RNA sequencing was employed to analyze transcriptional profiles of p190 CML (n=3) in contrast to p210 CML patients (n=4). A thorough transcriptional, phosphorylation and drug sensitivity profiling were applied to five p190- and three p210-expressing Ph+ALL patients. Expression alterations were further characterized in two cell line models mimicking BCR-ABL positive leukemia (Ba/F3 and HPCLSK). Phosphorylation profiles were analyzed by flowcytometry and phospho-array (Tyrosine Phosphorylation ProArray, Full Moon Biosystems). For drug sensitivity and resistance testing (DSRT), a custom plate set comprising 75 approved and investigational oncology drugs was used for patient samples and more extensive 528-drugs plates were used for the cell lines. Results: CML patients with p190 had a median age of 72.5 years at the diagnosis (range: 50-80) and all received imatinib as a frontline treatment. Only one patient achieved a fluctuating major molecular response (MMR) by 12 months while the rest of the patients showed primary resistance to treatment and were shifted to a 2nd line TKI, nilotinib (n=2) or proceeded to HSCT (n=1). By exome sequencing we identified 26 variants in p190-CML samples (median per patient=7, range: 2-10), including variants in ASXL1, DNMT3A and KDM4D genes. RNA-sequencing analysis identified 19 and 97 dysregulated genes (Q & lt;0.05) between p190- and p210 in CML and Ph+ ALL cells respectively. In CML, enrichment analysis revealed upregulation of TNF, interferon (IFN), IL1-R and Toll-like receptor (TLR) signaling, TP53-related, cell cycle and apoptosis pathways. Among Ph+ ALL samples, many CML-related genes were upregulated in samples encompassing p210 while IFN-, TP53- and cell cycle-related molecules were upregulated in p190 samples. p190 samples exhibited hyper-phosphorylation of Src kinase compared to p210 samples. DSRT results also revealed increased sensitivity of primary Ph+ ALL-p190 cells to Src-inhibitors (dasatinib and saracatinib), glucocorticoids and MDM2 inhibitors/TP53 activators (SAR405838 and idasanutlin). Regarding cell lines, Ba/F3-p190 showed the upregulation of interferon signaling pathways compared to p210. Src was also hyperphosphorylated in both Ba/F3 and HPCLSK p190 models. In addition to glucocorticoids and Src-inhibitors, compounds blocking the activity of the inhibitors of apoptosis protein (IAP) family were highly effective at reducing the viability of p190 compared to p210 cells in both cell lines. Conclusions: In CML, p190 isoform of BCR-ABL1 is associated with distinct features and should be considered as a high-risk group. Combining clinical, genomic, phosphorylation and drug sensitivity data, we demonstrated that p190 activates specific cancer pathways, notably Src signaling and interferon pathways. Data also suggests that CML patients with p190 could benefit from broad spectrum TKI with Src inhibiting activity or combination of TKI with MDM2- or IAP-inhibitors. Disclosures Heckman: Orion Pharma: Research Funding; Celgene: Research Funding; Novartis: Research Funding; Oncopeptides: Research Funding. Porkka:Celgene: Consultancy, Research Funding; Novartis: Consultancy, Research Funding; Daiichi Sankyo: Consultancy, Research Funding. Mustjoki:Novartis: Research Funding; Pfizer: Research Funding; BMS: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 136, No. Supplement 1 ( 2020-11-5), p. 4-5
    Abstract: Only half of chronic myeloid leukemia (CML) patients in deep molecular remission are able to maintain treatment free remission (TFR) after tyrosine kinase inhibitor (TKI) discontinuation. Identifying predictive markers for TFR remains a key issue. The immunological control or eradication of TKI insensitive leukemic stem cells may contribute to successful cessation, as recent work has demonstrated that differences in immune cell compositions can be associated with better probability of TFR. In order to understand the precise immunological changes in CML, we profiled over 170,000 cells from 25 CML samples from different clinical phases (untreated, during TKI therapy and after TKI cessation) using single-cell RNA and T cell receptor (TCR) αβ sequencing (scRNA+TCRab-seq). We profiled both CD45+ sorted peripheral blood samples (n=20) and CD34+ sorted bone marrow samples (n=5). To understand antigen-specific responses, we profiled TCRs specific to tumor-antigen PR1 (n=12) and compared these to unsorted TCRβ-sequenced samples from CML (n=137) and healthy donors (n=786). To understand the distinctive immunological features in CML, we compared the immune system in CML (n=20) to those in other hematological and solid malignancies profiled with scRNA+TCRab-seq (n=9). We discovered NK cells to be the most unique feature in CML, as NK CD56dim cells were more abundant in CML patients (Padj & lt;0.001) and the NK cell repertoire was more transcriptionally heterogenous and included otherwise rarely seen exhausted NK population with upregulated HAVCR2 and TIGIT expression (Padj & lt;0.0001). Further, the NK cell maturation changed during TKI-treatment from an effector CD56dim state towards an adaptive NK-cell state. We sought to understand whether the immunological activity differs before and after TKI discontinuation in different clinical outcomes following discontinuation. Therefore, we reclustered the discontinuation samples from three different outcomes: TFR, early relapse ( & lt;6 months after cessation) or late relapse ( & gt;6 months). In all clinical outcomes, TKI cessation invigorated NK cell exhaustion and the most upregulated pathways included IFNg and TNFa via NFkB pathways. Patients with successful TFR had less differences in their immune system after cessation in comparison to relapsing patients, which could hint that their immune subsets were better prepared for the cessation. Unlike the early relapse patients' quiescent immune cells, the late relapse patients' immune subsets were active, but under pronounced inhibitory Treg signals, providing a clinically interesting approach to alter the outcome of these patients. Next, to understand how malignant CML cells evade or interact with the immune system, we analyzed CD34+ cells and calculated a BCR-ABL1 activity score. The most primitive CD34+ cells had lower BCR-ABL1 score and not as many immunological ligand-receptor pairs as their less primitive, highly BCR-ABL1 pathway expressing, counterparts (P & lt;0.0001). LGALS9, that is expressed on both BCR-ABL1 high and low CD34+ cells, arose as one of the most interesting ligands mediating tumor-immune cell interactions and it has many potential receptors expressed by NK cells, including HAVCR2. Finally, we found that tumor-antigen PR1 is preferentially expressed on CD34+ cells with high BCR-ABL1 score. Most TCRs recognizing PR1 are restricted to individuals, but they share short amino-acid motifs that are shared across patients. Thus, we were able to generate an in silico tool to recognize PR1-specific TCRs from unsorted TCR-seq samples with high accuracy (AUC 0.91 from 10-fold cross validation). With our tool, we were able to show that anti-PR1 responses were more frequent in CML than in healthy donors (P & lt;0.0001), are more expanded in bone-marrow than in blood (P & lt;0.0001) and diversified during TKI treatment (P & lt;0.0001). In the scRNA+TCRab-seq data, the anti-PR1 response differed between TFR and early relapse patient significantly at phenotype level, where the TFR patient had cytotoxic anti-leukemic response before TKI cessation, in comparison to the early relapse patient's exhausted and less cytotoxic PR1-specific cells. In conclusion, our results provide high-resolution insights into anti-leukemic immune responses in CML and how we could harness and monitor them to enable successful TKI cessations. Figure Disclosures Olsson-Strömberg: Pfizer: Research Funding. Hjorth-Hansen:Pfizer: Honoraria, Research Funding; Austrian Orphan Pharma: Honoraria, Research Funding; Bristol-Myers Squibb: Research Funding. Mustjoki:Pfizer: Research Funding; Novartis: Research Funding; BMS: Honoraria, 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages