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: IBJ Plus, FUAM (Fundacioon Universidad Autonoma de Madrid)
    Type of Medium: Online Resource
    ISSN: 2531-0151
    Language: Unknown
    Publisher: FUAM (Fundacioon Universidad Autonoma de Madrid)
    Publication Date: 2018
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: JAMA Dermatology, American Medical Association (AMA), Vol. 152, No. 4 ( 2016-04-01), p. 435-
    Type of Medium: Online Resource
    ISSN: 2168-6068
    Language: English
    Publisher: American Medical Association (AMA)
    Publication Date: 2016
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 2719-2719
    Abstract: Advanced BCR-ABL1-positive leukemias (chronic myeloid leukemia in blast crisis and Ph+ALL) remain a therapy challenge despite advances in tyrosine kinase inhibitor (TKI) therapy. Emergence of primary and secondary resistance due to gatekeeper and compound mutations within the BCR-ABL1 kinase domain is common even with the novel 2nd and 3rd generation TKIs (dasatinib, nilotinib, ponatinib). We set out to identify novel candidate drugs for advanced BCR-ABL1-positive leukemias by using an unbiased high-throughput drug testing platform and utilizing both primary patient cells and cell lines. Methods As a study material we used 3 CML cell lines representing different types of CML blast phases. In addition to commonly used K562 cells, EM-2 and MOLM-1 cell lines were tested. AML cell lines (AML-193, AP-1060, HL60ATCC, HL60TB, Kasumi-1, KG-1, ME-1, MOLM-13, MONO-MAC-6, MUTZ-2, MV4-11, NOMO-1, SH-2, SHI-1, SIG-M5, SKM-1, THP-1) were used as cell line controls. To verify the results obtained from cell lines, primary bone marrow (BM) cells were derived from 2 TKI-resistant CML BC patients. Patient 1 had developed resistance to imatinib and dasatinib due to a T315I mutation, whereas patient 2 was resistant to nilotinib, dasatinib and ponatinib due to a V299L and a compound mutation. BM cells from 4 healthy individuals were used as controls. The functional profiling of drug responses was performed with a high-throughput drug sensitivity and resistance testing (DSRT) platform comprising of 306 anti-cancer agents (FDA/EMA approved, investigational and experimental compounds). Cells were dispensed to pre-drugged 386-well plates of 5 different concentrations and incubated in a humidified incubator with 5% CO2 at 37 °C for 72 hours. Cell viability was measured by using a luminescent cell viability assay (CellTiter-Glo). From plate reads a Drug Sensitivity Score (DSS) was calculated for each drug as a measure of cytotoxicity. In addition to DSRT, Human Phospho-Kinase Array Kit (R & D systems) was used to analyze the phosphokinase profile in patient samples. Results Based on initial comparisons between CML and AML cells lines, nonspecific cytotoxic drugs, which showed high activity in all cell lines, were omitted from further analysis. The DSS scores from different CML cells lines correlated relatively closely (EM-2 vs. K-562, r=0.89; EM-2 vs. MOLM-1, r=0.82; K-562 vs. MOLM-1, r=0.78; p 〈 0.0001 for all correlations). We next ranked the DSRT data according to the DSS values with most sensitive drugs showing the highest DSS scores. The primary cells from CML BC were further normalized against the median values from healthy controls, resulting in leukemia-specific sensitivity scores (sDSS). Ranked results from the DSRT analysis are shown in the Table. As expected, the cell lines were sensitive to TKIs, with the exception of the MOLM-1, which showed only modest sensitivity. The clinically TKI-resistant patient samples were also TKI-resistant ex vivo, further validating the DSRT assay data. Drugs which showed efficacy in both the cell lines and the TKI-resistant patients included HSP90 inhibitors (NVP-AUY922, BIIB021), a NAMPT inhibitor daporinad and the protein translation inhibitor omacetaxine (homoharringtonine). Phosphokinase antibody array results from the patient samples showed increased expression of the HSP27 protein as a putative biomarker for HSP90 inhibitor response. Conclusions DSRT is a powerful assay for identifying novel candidate molecules for refractory BCR-ABL1-positive leukemias. Our results indicate that HSP90 and NAMPT inhibitors in particular warrant further clinical evaluation both by analyzing a larger set of primary patient samples and by performing proof-of-concept clinical studies. The results also pave way for designing rational combination therapy strategies. Disclosures: Mustjoki: Novartis: Consultancy, Speakers Bureau; BMS: Consultancy, Speakers Bureau. Porkka:BMS: Consultancy, Research Funding, Speakers Bureau; Novartis: Consultancy, Research Funding, Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    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. 1725-1725
    Abstract: INTRODUCTION The recent success of checkpoint blockade immunotherapies in diverse solid tumors has prompted the evaluation of these treatments in hematologic malignancies such as acute myeloid leukemia (AML). It is critical to identify the patient and disease subsets that could respond to such therapies. Infiltration of tumors by cytotoxic T lymphocytes (CTLs) has been associated with better prognosis and responses to checkpoint inhibition. We hypothesized that the presence of a substantial fraction of activated CTLs and natural killer (NK) cells in the blood and bone marrow samples of hematologic tumors could indicate a preexisting active immune response potentially targeting the tumor cells. Moreover, the density of the immune infiltrate could shape and be shaped by the expression of cancer-germline and leukemia-associated antigens (LAAs), antigen-presenting machinery (APM) and immunosuppressive genes by the tumor cells. Here, we examined these immunological properties of hematological tumors in large-scale gene expression datasets to identify immunologically active patient subsets. METHODS Curated set of 9,544 transcriptomes collected across 36 hematological malignancies (HEMAP), including 1,858 AML cases was utilized to identify subsets of patients with existing, potentially tumor-directed immune responses. Additional multi-omics datasets of 173 AML patients from The Cancer Genome Atlas (TCGA) were integrated to gain insight into the genetic landscape of immunologically active patients. Cytolytic activity (geometric mean of GZMA (granzyme A) and PRF1 (perforin) transcript levels, Rooney et al., Cell 2015) was used as a marker of immunologic activity. Cytolytic activity was correlated to the expression levels of all transcripts, gene sets from collections such as MSigDB and manually curated gene sets representing the APM (HLA-A, -B, -C, B2M), 145 known cancer-germline antigens as well as established LAAs such as WT1 and PRTN3. Furthermore, we used an in silico flow cytometry approach, CIBERSORT (Newman et al., Nat Methods 2015), to infer the relative fractions of 22 immune cell subpopulations from the gene expression data to dissect the immune cell composition of the samples. RESULTS Cytolytic activity showed high correlation with other transcripts expressed in activated CTLs and NK cells (e.g. GZMB, GNLY, KLRB1, CD8A, CD2; Spearman's R ≥ 0.7) as well as lymphocyte activation-related gene sets across both the HEMAP and the TCGA AML datasets, validating it as a robust and specific metric of active cellular immunity. When correlated to the CIBERSORT immune cell populations, cytolytic activity was positively associated with CD8 T cells and showed a negative correlation to the proportion of M2 macrophages. High levels of cancer-germline antigens were associated with decreased expression of components of the APM and low cytolytic activity, suggesting HLA downregulation as a mechanism of immune evasion by cancer-germline antigen-expressing tumor cells. We observed extensive heterogeneity in the cytolytic activity between different diseases and subtypes within the same disease, most prominently in AML. In AML patients, complex karyotype and unfavorable prognosis were correlated with high cytolytic activity, indicating biological similarity of the immune-infiltrated tumors. Furthermore, TP53 mutations, genome fragmentation and immune checkpoint transcripts such as CD274 (PD-L1), PDCD1LG2 (PD-L2), CTLA4 and LAG3 were enriched within the complex karyotype cluster in the TCGA AML dataset. In contrast, mutations in NPM1 and FLT3 showed a modest but significant negative correlation to cytolytic activity. CIBERSORT analysis revealed that AML cases with low cytolytic activity preferentially had enrichment of an eosinophilic phenotype in addition to increased M2 polarization of macrophages. CONCLUSIONS Using large-scale transcriptomics approaches, we were able to identify patient subsets with variable levels of immune cytolytic activity within hematologic malignancies. Furthermore, we identified connections between the cytotoxic immune response and genetic properties of AML tumors. These observations have potential clinical implications, as the choice of patients to clinical trials receiving immune checkpoint blockade immunotherapies would require careful consideration in light of the observed immunological heterogeneity. Disclosures Mustjoki: Bristol-Myers Squibb: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Ariad: 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. 126, No. 23 ( 2015-12-03), p. 700-700
    Abstract: Background Natural killer (NK) cell malignancies are rare lymphoid neoplasms characterized by aggressive clinical behavior and poor treatment outcomes. Clinically they are classified as extranodal NK/T-cell lymphoma, nasal type (NKTCL) and aggressive NK cell leukemia (ANKL). Both subtypes are almost invariably associated with Epstein-Barr virus (EBV). Recently, genomic studies in NKTCL have identified recurrent somatic mutations in JAK-STAT pathway molecules STAT3 and STAT5b as well as in the RNA helicase gene DDX3X in addition to previously detected chromosomal aberrations. Here, we identified somatic mutations in 4 cases of ANKL in order to understand whether these entities share common alterations at the molecular level. To further establish common patterns of deregulated oncogenic signaling pathways operating in malignant NK cells, we performed drug sensitivity profiling using NK cell lines representing ANKL, NKTCL and other malignant NK cell proliferations. We aimed to identify sensitivities to agents that selectively target components of pathways required for survival of malignant NK cells in an unbiased manner. Methods Exome sequencing was performed on peripheral blood or bone marrow of ANKL patients using the NK cell negative fraction or other healthy tissue as control. Profiling of drug responses was performed with a high-throughput drug sensitivity and resistance testing (DSRT) platform comprising 461 approved and investigational oncology drugs. The NK cell lines KAI3, KHYG-1, NKL, NK-YS, NK-92, SNK-6 and YT and IL-2-stimulated and resting NK cells from healthy donors were used as sample material. All drugs were tested on a 384-well format in 5 different concentrations over a 10,000-fold concentration range for 72 h and cell viability was measured. A Drug Sensitivity Score (DSS) was calculated for each drug using normalized dose response curve values. Results The ANKL patients displayed mutations in genes reported as recurrently mutated in NKTCL, such as FAS, TP53, NRAS, STAT3 and DDX3X. Additionally, novel alterations in genes previously implicated in the pathogenesis of NKTCL were detected. These included an inactivating mutation in INPP5D (SHIP), a negative regulator of the PI3K/mTOR pathway and a missense mutation in PTPRK, a negative regulator of STAT3 activation. Interestingly, the total number of nonsilent somatic mutations in 3 out of 4 ANKL patients (97, 82 and 45) was remarkably high compared to other hematological malignancies analyzed in our variant calling pipeline. Analysis of drug sensitivities in NK cell lines showed a close correlation between all cell lines and a markedly higher correlation with those of IL-2 stimulated than resting healthy NK cells, suggesting that malignant NK cells may share a common drug response pattern. Furthermore, in an unsupervised hierarchical clustering the NK cell lines formed a distinct group from other leukemia cell lines tested (Fig. A). Among pathway-selective compounds (namely, kinase inhibitors and rapalogs), the drugs most selective for malignant NK cells fell into two major categories: PI3K/mTOR inhibitors (e.g. temsirolimus, buparlisib) and inhibitors of aurora and polo-like kinases such as rigosertib and GSK-461364 (Fig. B). JAK inhibitors (e.g. ruxolitinib, gandotinib) and CDK inhibitors (e.g. dinaciclib) showed strong efficacy in both malignant NK cells and IL-2 activated healthy NK cells. Conclusions Our exome sequencing results suggest that candidate driver alterations affecting similar signaling pathways underlie the pathogenesis of ANKL as has been reported in NKTCL. Drug sensitivity profiling highlights the PI3K/mTOR pathway as a potential major driver of malignant NK cell proliferation, whereas JAK-STAT signaling appears to be essential in both healthy and malignant NK cells. Components of these pathways harbored mutations in our small cohort of ANKL patients and have been shown to be deregulated by mutations or other mechanisms in previous studies, underlining their importance as putative drivers. The systematic large-scale characterization of drug responses also identified these pathways as potential targets for novel therapy strategies in NK cell malignancies. Figure 1. (A) Unsupervised hierarchical clustering based on drug sensitivity scores (DSS) of NK, AML, CML and T-ALL cell lines. (B) Scatter plot comparing DSS of malignant NK cell lines (average) and healthy IL-2 stimulated NK cells. Figure 1. (A) Unsupervised hierarchical clustering based on drug sensitivity scores (DSS) of NK, AML, CML and T-ALL cell lines. (B) Scatter plot comparing DSS of malignant NK cell lines (average) and healthy IL-2 stimulated NK cells. Disclosures Mustjoki: Novartis: Honoraria, Research Funding; Bristol-Myers Squibb: Honoraria, Research Funding; Pfizer: Honoraria, 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 ...
  • 6
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 3099-3099
    Abstract: Predicting minimal residual disease (MRD) levels in tyrosine kinase inhibitor (TKI)-treated chronic myeloid leukemia (CML) patients is of major clinical relevance. The reason is that residual leukemic (stem) cells are the source for both, potential relapses of the leukemicclone but also for its clonal evolution and, therefore, for the occurrence of resistance. The state-of-the art method for monitoring MRD in TKI-treated CML is the quantification of BCR-ABL levels in the peripheral blood (PB) by PCR. However, the question is whether BCR-ABL levels in the PB can be used as a reliable estimate for residual leukemic cells at the level of hematopoietic stem cells in the bone marrow (BM). Moreover, once the BCR-ABL levels have been reduced to undetectable levels, information on treatment kinetics is censored by the PCR detection limit. Clearly, BCR-ABL negativity in the PB suggests very low levels of residual disease also in the BM, but whether the MRD level remains at a constant level or decreases further cannot be read from the BCR-ABL negativity itself. Thus, also the prediction of a suitable time point for treatment cessation based on residual disease levels cannot be obtained from PCR monitoring in the PB and currently remains a heuristic decision. To overcome the current lack of a suitable biomarker for residual disease levels in the BM, we propose the application of a computational approach to quantitatively describe and predict long-term BCR-ABL levels. The underlying mathematical model has previously been validated by the comparison to more than 500 long-term BCR-ABL kinetics in the PB from different clinical trials under continuous TKI-treatment [1,2,3]. Here, we present results that show how this computational approach can be used to estimate MRD levels in the BM based on the measurements in the PB. Our results demonstrate that the mathematical model can quantitatively reproduce the cumulative incidence of the loss of deep and major molecular response in a population of patients, as published by Mahon et al. [4] and Rousselot et al. [5] . Furthermore, to demonstrate how the model can be used to predict the BCR-ABL levels and to estimate the molecular relapse probability of individual patients, we compare simulation results with more than 70 individual BCR-ABL-kinetics. For this analysis we use patient data from different clinical studies (e.g. EURO-SKI: NCT01596114, STIM(s): NCT00478985, NCT01343173) where TKI-treatment had been stopped after prolonged deep molecular response periods. Specifically, we propose to combine statistical (non-linear regression) and mechanistic (agent-based) modelling techniques, which allows us to quantify the reliability of model predictions by confidence regions based on the quality (i.e. number and variance) of the clinical measurements and on the particular kinetic response characteristics of individual patients. The proposed approach has the potential to support clinical decision making because it provides quantitative, patient-specific predictions of the treatment response together with a confidence measure, which allows to judge the amount of information that is provided by the theoretical prediction. References [1] Roeder et al. (2006) Dynamic modeling of imatinib-treated chronic myeloid leukemia: functional insights and clinical implications, Nat Med 12(10):1181-4 [2] Horn et al. (2013) Model-based decision rules reduce the risk of molecular relapse after cessation of tyrosine kinase inhibitor therapy in chronic myeloid leukemia, Blood 121(2):378-84. [3] Glauche et al. (2014) Model-Based Characterization of the Molecular Response Dynamics of Tyrosine Kinase Inhibitor (TKI)-Treated CML Patients a Comparison of Imatinib and Dasatinib First-Line Therapy, Blood 124:4562 [4] Mahon et al. (2010) Discontinuation of imatinib in patients with chronic myeloid leukaemia who have maintained complete molecular remission for at least 2 years: the prospective, multicentre Stop Imatinib (STIM) trial. Lancet Oncol 11(11):1029-35 [5] Rousselot 
et al. (2014) Loss of major molecular response as a trigger for restarting TKI therapy in patients with CP- CML who have stopped Imatinib after durable undetectable disease, JCO 32(5):424-431 Disclosures Glauche: Bristol Meyer Squib: Research Funding. von Bubnoff:Amgen: Honoraria; Novartis: Honoraria, Research Funding; BMS: Honoraria. Saussele:ARIAD: Honoraria; Novartis: Honoraria, Other: Travel grants, Research Funding; Pfizer: Honoraria, Other: Travel grants; BMS: Honoraria, Other: Travel grants, Research Funding. Mustjoki:Bristol-Myers Squibb: Honoraria, Research Funding; Pfizer: Honoraria, Research Funding; Ariad: Research Funding; Novartis: Honoraria, Research Funding. Guilhot:CELEGENE: Consultancy. Mahon:NOVARTIS PHARMA: Honoraria, Research Funding; BMS: Honoraria; PFIZER: Honoraria; ARIAD: Honoraria. Roeder: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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 477-477
    Abstract: Rationale: Dasatinib (DAS) and interferon have different modes of action and may have synergistic activity in CML, due to both antineoplastic and immunostimulatory mechanisms. Addition of pegylated interferon (PegIFN) to imatinib therapy in CP-CML has in previous clinical trials (French SPIRIT and NordCML002) resulted in deeper molecular responses. Thus, an optimal combination of DAS and PegIFN may increase the proportion of patients who reach deep molecular response with potential for treatment-free remission (TFR). Design: Newly diagnosed CP-CML patients were treated with DAS (Sprycel, BMS) 100 mg OD as single drug for three months. Thereafter weekly subcutaneous injections of Peg-IFN α2b (PegIntron, MSD) were added to DAS; from end of month 3 (M3) to M6, 15µg/week, thereafter 25µg/week until M15. Primary end points were safety and the rate of MMR at M12. The doses of PegIFN were lower than in the SPIRIT and NordCML002 studies to increase adherence. Population: Forty patients were included at 14 university centers. One patient was lost to follow-up after M6. All patients were included in analysis up to M12. Mean and median age was 48 years (range 19-71). The proportions of high risk patients were 25% (Sokal), 15% (Hasford), and 15% (EUTOS). Safety and dosing: Treatment was well tolerated with expected DAS and PegIFN related side effects. Six patients had seven serious adverse events (AEs), all hospitalizations. 1 episode each of bradycardia/atrial fibrillation (possibly PegIFN-related), headache (DAS), fever (PegIFN), anaphylaxis-like reaction (PegIFN), fever/malaise/headache (PegIFN), pneumonia and a knee effusion (both unrelated). One pleural effusion occurred (grade 2, 3%). Grade 3-4 neutropenia and thrombocytopenia occurred in 6 and 9 patients respectively. Prolonged hematological toxicity ( 〉 2 months) occurred in 8 patients, causing dosing problems in 5. One patient suffered grade 3 depression. Grade 3 flu-like symptoms occurred in 2 patients. One patient had lipase elevation grade 3 and one patient developed hypothyroidism attributed to PegIFN. Grade 2 dermal AEs like rash and acne occurred in about 20%, attributable to both drugs. 94% (DAS) and 76% (PegIFN) of assigned dose was given. Dose reductions occurred in 19 patients for DAS and 20 patients for PegIFN. Two patients discontinued DAS and switched to nilotinib, 1 for headache at M3 and 1 at M12 for lack of efficacy/hematological toxicity. Two patients could not start PegIFN for hematological toxicity (one lost to follow-up after M6). PegIFN was discontinued because of bradycardia/atrial fibrillation (1 patient), anaphylaxis (1 patient), flu-like syndrome (2 patients) and long-term hematological toxicity (2 patients). At 12 months 31/38 pats (82%) were still on PegIFN, a higher proportion than in the French Spirit or NordCML002 studies. Efficacy: We have used the DAS arm of the Dasision study (Kantarjian NEJM 2010) as a historical control. Early response at M3 was very similar between studies. In the present and the Dasision cohorts respectively, 18% vs 16% missed the 10% BCR-ABLIS landmark, 66% vs 56% achieved a CCyR and 8% vs 8% achieved MMR. At M6, three months after introduction of PegIFN, a steep increase in MMR rate was observed compared with Dasision. This was also reflected in deep responses, MR4.0 (see tables) and MR4.5 at M12, 18% vs 5%. The primary efficacy endpoint was MMR at M12, 82% vs 46%. Table 1.MMRDAS+PegIFN (%)DAS (Dasision)(%)Difference (%)M3880M6532726M9663927M12824636Table 2.MR4.0DAS+PegIFN (%)DAS (Dasision) (%)Difference (%)M3303M620614M938830M12481236 Progressions and treatment failure defined by ELN 2013: Failures: No progression was noted. At M3, 2 patients still had 〉 95% Ph+ metaphases (MF). At M6, four patients (11%) had 〉 35% Ph+MF or 〉 10% BCR-ABL levels. At M12, one patient failed CCgR and two more patients failed 〈 1% BCR-ABL. No BCR-ABL mutations were detected in "failure" patients. Conclusion: The combination of DAS and low dose PegIFN could be safely administered in newly diagnosed CP CML. No unexpected autoimmune phenomena were observed, and pleural effusions were rare. Efficacy appears very promising with high early MMR rates and deep molecular responses. A randomized comparison DAS +/- PegIFN is warranted. Support: Study drug from BMS and MSD. Grant from BMS. Figure 1. Figure 1. Disclosures Hjorth-Hansen: Ariad: Honoraria; Novartis: Honoraria; Pfizer: Honoraria, Research Funding; Bristol-Myers Squibb: Research Funding. Off Label Use: Dasatinib and Pegylated IFN combination in CML. Richter:Ariad: Honoraria; Bristol-Myers Squibb: Honoraria; Novartis: Honoraria. Porkka:Bristol-Myers Squibb: Honoraria; Celgene: Honoraria; Novartis: Honoraria; Pfizer: Honoraria. Mustjoki: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: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 2782-2782
    Abstract: Abstract 2782 Background: The inhibition of oncogenic BCR-ABL1 kinase with tyrosine kinase inhibitors (TKIs) has significantly improved the prognosis of CML, but the majority of patients need life-long therapy. Before the TKI era, CML patients were treated with interferon-α (IFN-α), and a minor proportion of patients (10–20%) achieved prolonged complete cytogenetic remissions (CCyR). Interestingly, many of the patients in prolonged CCyR were able to discontinue the treatment without imminent disease relapse. As the ultimate goal of current CML therapy is cure, the deeper understanding of the cellular mechanisms behind successful IFN-α discontinuation are of outmost importance. In this study, we analyzed the function of immune effector cells (NK- and T-cells) derived from CML patients in prolonged molecular remission after IFN-α monotherapy in order to understand the putative anti-leukemic effects. METHODS: The study cohort included 13 CML patients treated with IFN-α who were in complete molecular remission (CMR) at the time of blood sampling. Five patients were still on IFN-α therapy (IFN-ON, median duration of the treatment 13.7 years) and 8 had stopped treatment successfully (IFN-OFF) and were off from any treatment (median time without treatment 6.3 years). None of the patients had used any TKIs. Samples from 10 healthy volunteers were used as controls. The cytotoxicity of NK-cells in blood was studied by measuring the direct killing of K562 cells and by degranulation assay (CD107). The cytokine secretion (TNF-α and IFN-g) of NK-cells was measured with flow cytometry after stimulation with K562. The function of T-cells was studied by antibody (OKT3) stimulation and measuring TNF-α and IFN-g production by flow cytometry. In addition, NK-cells were phenotyped by multi-color flow cytometry. RESULTS: IFN-OFF patients had a larger proportion of NK-cells from lymphocytes than IFN-ON patients or healthy controls (IFN-OFF median 23.5 %, IFN-ON 7.0 %, healthy 13.2 %; p=0.0136). Based on the subset analysis, expanded NK-cells in IFN-OFF patients had mature phenotype CD56dimCD62LlowCD27lowCD57high. On the contrary, IFN-ON patients had a larger proportion of immunoregulatory CD56bright NK-cells (20.2% vs. 6.5% in healthy and 3.0% in IFN-OFF, p=0.0035). When the direct killing of K562 cells was studied, NK-cells from healthy controls killed better than NK-cells from either of the patients groups (in healthy 55 % of K562 cells were alive compared to 79% in IFN-ON and 91% in IFN-OFF, p=0.0042). Similar trend was also observed in degranulation assay (median degranulation in healthy controls 8.3 % compared to 4.3 % in IFN-OFF and 3.6 % in IFN-ON group, p= 0.39). The direct killing capability tended to correlate negatively r=-0.60, p=0.07) with the NK-cell proportion (ie. patients with high NK-cell counts had worse killing). Instead of cytotoxicity, NK-cells from IFN-OFF patients seemed to secrete more efficiently cytokines (IFN-γ/TNF-α) than NK-cells from healthy controls, but due to low number of patients no firm conclusions can be made (IFN-OFF median 6.9 %, healthy 0.7 %; p= 0.25). The proportion of potentially cytotoxic CD4+GrB+ T-cells did not significantly differ between the groups, but wide variation was observed between the individual patients (IFN-ON median 9.0 %, IFN-OFF 2.6 %, and healthy controls 3.5 %; p=0.4142). However, the secretion of TNF-α and IFN-γ by CD4+ cells seemed to be increased in IFN-OFF patients when compared to healthy controls (IFN-OFF mean 5.9 %, IFN-ON 3.7 %, healthy 2.5 %; p=0.1055). CONCLUSIONS: CML patients who have been able to discontinue IFN-α therapy (IFN-OFF group) had an expansion of mature CD56dimCD62LlowCD27lowCD57high NK-cells in peripheral blood. Although CD56dim NK-cells have typically been suggested to act as cytotoxic cells, the killing in response to K562 cells was lower in IFN-OFF patients than in controls. It is possible that the expanded NK-cells in IFN-OFF patients represent exhausted terminal stage cells (CD57+) or memory cells that do not react properly against third party target cells (K562). Moreover, they may have an immunoregulatory function instead of cytotoxicity. Further follow-up studies with other patient cohorts (TKI monotherapy and TKI+IFN-α combination therapy treated patients) are ongoing to clarify the issue further. Disclosures: Porkka: Bristol-Myers Squibb: Honoraria, Research Funding; Novartis: Honoraria, Research Funding. Mustjoki:Bristol-Myers Squibb: Honoraria, Research Funding; Novartis: Honoraria.
    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 ...
  • 10
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 3600-3600
    Abstract: In the past two decades spontaneous erythroid (BFU-E) and megakaryocytic (CFU-Meg) colony formation have proved to be useful diagnostic tools in diagnosing myeloproliferative disorders (MPD). Recently a point mutation in the JAK2-gene was discovered to be a pathogenetic event in polycythemia vera (PV) and essential thrombocythemia (ET). JAK2 mutation analysis has been recommended to be used as a primary diagnostic method for these disorders. However, so far only a few studies comparing the in vitro growth pattern of hematopoietic progenitors and JAK2 mutation status have been published and, to the best of our knowledge, no studies describing the association between spontaneous CFU-Meg growth and the JAK2 status have been conducted. Therefore, the aim of this study was to compare the results obtained with these methods in PV and ET. 52 ET and 33 PV patients were studied. Allele-specific PCR based JAK2 mutation analysis and hematopoietic colony forming assays were done from bone marrow (BM) aspirate samples. Morphology of BM aspirates was analyzed in our routine diagnostic laboratory. 30/33 (91%) PV patients and 35/52 (67%) ET patients showed spontaneous BFU-E growth. Spontaneous CFU-Meg growth was found in 23/33 (70%) of PV patients and in 29/52 (56%) of ET patients. JAK2 mutation was seen in 26 (79%) PV patients and in 31 (60 %) ET patients. All JAK2 mutated PV and ET patients were found to have spontaneous BFU-E growth. In addition, 4 of 7 (57%) JAK2 mutation negative PV patients and 4 of 21 (19%) JAK2 mutation negative ET patients had spontaneous BFU-E colony formation. JAK2 mutated ET patients had spontaneous CFU-Meg growth more often than JAK2 mutation negative patients (71% vs. 33%), while in PV patients there was no clear difference between the two groups (JAK2 mutation positive 69% vs. negative 71%). Interestingly, 9 patients (6 with ET, 3 with PV) had only spontaneous CFU-Meg growth but no spontaneous BFU-E growth. They were all JAK2 mutation negative. In addition, 11/52 ET patients (21%) were JAK2 mutation negative and had neither spontaneous BFU-E nor spontaneous CFU-Meg growth. BM morphology was considered suggestive of ET or PV in 83% and 70% of the cases respectively. No significant differences in BM morphology were found between JAK2 mutation negative and positive patients. In conclusion, in this group of 88 MPD patients spontaneous BFU-E colony growth was the most sensitive diagnostic assay. All patients with JAK2 mutation also had spontaneous BFU-E growth and an additional 8 patients without the mutation also had spontaneous BFU-E growth. Although recently the JAK2 mutation has been described in megakaryocytes as well, none of the present patients with spontaneous CFU-Meg growth but without spontaneous BFU-E growth were JAK2 mutation positive. This suggests that JAK2 mutation may not be the only pathogenetic event causing spontaneous CFU-Meg growth and further studies are needed to define genetic alterations behind this phenomenon.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2006
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    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...