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
  • 1
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 20, No. 6 ( 2014-03-15), p. 1576-1589
    Abstract: Background: Stromal-mediated signaling enhances NF-κB pathway activity in chronic lymphocytic leukemia (CLL) B cells, leading to cell survival and chemoresistance. Ubiquitination of IκBα may partially account for constitutive activation of NF-κB. MLN4924 is an investigational agent that inhibits the Nedd8-activating enzyme, thereby neutralizing Cullin-RING ubiquitin ligases and preventing degradation of their substrates. Experimental Design: We conducted a preclinical assessment of MLN4924 in CLL. Primary CLL cells were cocultured in vitro with CD40L-expressing stroma to mimic the prosurvival conditions present in lymphoid tissue. The effect of MLN4924 on CLL cell apoptosis, NF-κB pathway activity, Bcl-2 family members, and cell cycle was assessed by flow cytometry, Western blotting, PCR, and immunocytochemistry. Results: CD40L-expressing stroma protected CLL cells from spontaneous apoptosis and induced resistance to multiple drugs, accompanied by NF-κB activation and Bim repression. Treatment with MLN4924 induced CLL cell apoptosis and circumvented stroma-mediated resistance. This was accompanied by accumulation of phospho-IκBα, decreased nuclear translocation of p65 and p52 leading to inhibition of both the canonical and noncanonical NF-κB pathways, and reduced transcription of their target genes, notably chemokines. MLN4924 promoted induction of Bim and Noxa in the CLL cells leading to rebalancing of Bcl-2 family members toward the proapoptotic BH3-only proteins. siRNA-mediated knockdown of Bim or Noxa decreased sensitivity to MLN4924. MLN4924 enhanced the antitumor activity of the inhibitors of B-cell receptor (BCR)–associated kinases. Conclusions: MLN4924 disrupts NF-κB activation and induces Bim expression in CLL cells, thereby preventing stroma-mediated resistance. Our data provide rationale for further evaluation of MLN4924 in CLL. Clin Cancer Res; 20(6); 1576–89. ©2014 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: PLOS ONE, Public Library of Science (PLoS), Vol. 10, No. 11 ( 2015-11-25), p. e0143685-
    Type of Medium: Online Resource
    ISSN: 1932-6203
    Language: English
    Publisher: Public Library of Science (PLoS)
    Publication Date: 2015
    detail.hit.zdb_id: 2267670-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Experimental Hematology, Elsevier BV, Vol. 38, No. 12 ( 2010-12), p. 1167-1177
    Type of Medium: Online Resource
    ISSN: 0301-472X
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2010
    detail.hit.zdb_id: 2005403-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Leukemia & Lymphoma, Informa UK Limited, Vol. 56, No. 5 ( 2015-05-04), p. 1566-1569
    Type of Medium: Online Resource
    ISSN: 1042-8194 , 1029-2403
    Language: English
    Publisher: Informa UK Limited
    Publication Date: 2015
    detail.hit.zdb_id: 2030637-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Society of Hematology ; 2013
    In:  Blood Vol. 122, No. 21 ( 2013-11-15), p. 1636-1636
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 1636-1636
    Abstract: Cyclins are important regulators of cell cycle in neoplastic cells. Regimented fluctuation in their expression is required for orderly cell cycle progression. Cyclins partner and modulate cyclin-dependent kinases (CDK) which are necessary for their activity. Since neoplastic cells are characterized by deregulated cell cycle with excessive proliferation, targeting cell cycle progression has been an attractive therapeutic approach. CDK inhibitors, most notably flavopiridol and dinaciclib, have shown efficacy in early phase clinical trials in CLL. The dominant fraction of the CLL clone is represented by B-cells in a resting (G0) state of the cell cycle. Efficacy of the CDK inhibitors in CLL is attributed to their effect on CDK-7/9 which facilitate initiation and sustenance of RNA polymerase-mediated transcription. P1446A is a novel potent and orally active kinase inhibitor which inhibits CDK's at nanomolar concentrations (CDK-1 IC50 25 nM, CDK-4 IC50 90 nM, CDK-6 IC50 210 nM and CDK-9 IC5022 nM). P1446A has shown clinical activity in Phase I studies in solid tumors and exhibited minimal hematologic toxicity. Here we studied activity of P1446 in primary CLL B-cells and MEC1 cells. We enrolled 40 patients with median age of 70 years at the CLL clinic at Dartmouth. Median time of follow-up was 4 years and most patient (75%) were untreated. 60% of patients had unmutated IGHV. The majority of patients had normal cytogenetics, trisomy 12 or 13q deletion, and 3 patients (10%) had deletion 11q. Additionally, 10 patients with deletion 17p were enrolled at the CLL Center at Dana Farber Cancer Institute. CLL B cells were isolated from peripheral blood using standard Ficoll-Hypaque technique and cultured in RPMI supplemented with 15% fetal bovine serum. Apoptosis was quantified by means of Annexin V/7-AAD staining and flow cytometry as well as immunoblotting (PARP cleavage). B-cell receptor simulation was performed with 10 mg/mL immobilized anti-IgM. We also determined activity of P1446 in MEC1 cells. CLL cells showed features of apoptosis (PARP cleavage) after a 2-hour incubation with 1500 nM P1446A. While apoptosis began to occur with lower concentrations of P1446A (0.2 μM), maximal efficacy was reached with 1.5 μM. After 24 hours of incubation with that dose, 84.1±4.7% CLL cells demonstrated Annexin V staining, compared to 30.2±2.5% of cells treated with vehicle control (p 〈 0.001). P1446A-induced cell death did not depend on the IGHV mutational status (p=0.44) or cytogenetic abnormalities. However, samples which carried deletion 17p showed decreased sensitivity towards the CDK inhibitor compared with those which had normal cytogenetics or non-17p abnormality (p=0.003). Still, 37.5±4.4% cells carrying 17p del underwent apoptosis upon 24-hour exposure to 1.5 μM P1446 (normalized to untreated control). P1446A was significantly more toxic to CLL cells compared to normal B-cells (p 〈 0.001). To determine whether P1446A had an impact on BCR signaling-mediated survival, we co-cultured CLL cells with 10 mg/mL immobilized anti-IgM. As expected, BCR engagement resulted in CLL cell rescue from spontaneous apoptosis in a subset of patient samples, while co-incubation with P1446A abrogated this protective effect. Similarly, P1446 induced significant apoptosis in MEC1 cell line leading to apoptosis induction after 6 hours of incubation with 1.5 μM P1446A. We determined that 48-hour incubation with 3 μM P1446A resulted in a two-fold reduction of MEC1 metabolic activity in an MTT assay. Our data shows that a novel CDK inhibitor P1446A demonstrates significant pre-clinical activity in CLL. Disclosures: Agarwal: Piramal Enterprizes Ltd.: Employment.
    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 ...
  • 6
    In: British Journal of Haematology, Wiley, Vol. 178, No. 5 ( 2017-09), p. 820-823
    Type of Medium: Online Resource
    ISSN: 0007-1048 , 1365-2141
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2017
    detail.hit.zdb_id: 1475751-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood Cancer Discovery, American Association for Cancer Research (AACR), Vol. 4, No. 3 ( 2023-05-01), p. 228-245
    Abstract: RNA splicing dysregulation underlies the onset and progression of cancers. In chronic lymphocytic leukemia (CLL), spliceosome mutations leading to aberrant splicing occur in ∼20% of patients. However, the mechanism for splicing defects in spliceosome-unmutated CLL cases remains elusive. Through an integrative transcriptomic and proteomic analysis, we discover that proteins involved in RNA splicing are posttranscriptionally upregulated in CLL cells, resulting in splicing dysregulation. The abundance of splicing complexes is an independent risk factor for poor prognosis. Moreover, increased splicing factor expression is highly correlated with the abundance of METTL3, an RNA methyltransferase that deposits N6-methyladenosine (m6A) on mRNA. METTL3 is essential for cell growth in vitro and in vivo and controls splicing factor protein expression in a methyltransferase-dependent manner through m6A modification-mediated ribosome recycling and decoding. Our results uncover METTL3-mediated m6A modification as a novel regulatory axis in driving splicing dysregulation and contributing to aggressive CLL. Significance: METTL3 controls widespread splicing factor abundance via translational control of m6A-modified mRNA, contributes to RNA splicing dysregulation and disease progression in CLL, and serves as a potential therapeutic target in aggressive CLL. See related commentary by Janin and Esteller, p. 176. This article is highlighted in the In This Issue feature, p. 171
    Type of Medium: Online Resource
    ISSN: 2643-3230 , 2643-3249
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 2875-2875
    Abstract: Gene expression profiling identified lymph nodes as the site of cell activation and proliferation with upregulation of NFκB. B-cell receptor (BCR) and CD40 signaling enhances NFκB pathway activity in CLL cells resident in the microenvironment, leading to their survival, proliferation and chemoresistance. The NFκB pathway has been successfully targeted in CLL in vitro, yet lack of clinical advances with those agents necessitates development of novel approaches. Ubiquitination of IκBα may partially account for constitutive activation of NFκB in this setting. MLN4924 is an investigational agent that inhibits the Nedd8-activating enzyme and thereby prevents neddylation of Cullin-RING ubiquitin ligases, resulting in a decrease of their activity and stabilization of their protein substrates. This study reports that MLN4924 abrogates pro-survival microenvironment stimuli in CLL cells in vitro. Primary B cells from 50 patients with CLL were co-cultured in vitro with CD40L-expressing stroma to mimic the pro-survival conditions present in lymphoid tissue. The effect of MLN4924 on CLL cell apoptosis, NFκB pathway activity, Bcl-2 family members and cell cycle was assessed by flow cytometry, western blotting, PCR and immunocytochemistry. MLN4924 was provided by Millennium Pharmaceuticals. Treatment with MLN4924 led to reduced neddylation of Cullin-RING ubiquitin ligases, accumulation of phospho-IκBa and induced modest apoptosis in CLL cells cultured off stroma (12.8±1.7% after a 24 hour incubation with 1 μM MLN4924 compared to untreated control). Incubation with the CD40L-expressing stroma resulted in activation of the canonical and non-canonical NFκB pathways in the CLL cells, induction of Mcl-1 and Bcl-xL, leading to protection from spontaneous apoptosis. This was accompanied by repression of Bim, a pro-apoptotic BH3-only protein of the Bcl-2 family. CD40L-expressing stroma rescued CLL cell from drug-induced apoptosis, including bendamustine, fludarabine, chlorambucil and CAL-101. By contrast, CD40L-expressing stroma failed to elicit resistance to MLN4924. Moreover, CLL cells cultured on stroma were more sensitive to Nedd8-activating enzyme inhibition compared to cells off stroma (1 μM MLN4924 induced apoptosis in 55.3±3.4% cells cultured on stroma). MLN4924 promoted apoptosis regardless of the common adverse prognostic factors (IGHV mutational status, cytogenetic markers, or ZAP-70 expression). In the presence of MLN4924, CLL cells demonstrated a significant decrease in nuclear translocation of the NFκB subunits p65 and p52 thus marking de-activation of both canonical and non-canonical pathways. Using gene microarray analysis we determined that receptor signaling and expression target pathways involving NFκB were most significantly associated with genes downregulated in CLL cells by MLN4924 (p 〈 0.0001). We noted reduced transcription of several groups of NFκB target genes, including anti-apoptotic Bcl-2 family members and genes involved in cell cycle progression (p 〈 0.01). Furthermore, treatment with MLN4924 resulted in a significant downregulation of a number of important cytokine ligands and receptors expressed by the CLL cells, including CCL5, CCL22, CXCR7, CXCR5 and CD40. Concomitantly, MLN4924 promoted induction of Bim and Noxa in the CLL cells which preceded apoptosis as evidenced by PARP cleavage. siRNA-mediated knockdown of either Bim or Noxa resulted in partial protection from MLN4924-mediated apoptosis irrespective of expression of the pro-survival Bcl-2 family members (Bcl-2, Bcl-xL or Mcl-1). This indicates that MLN4924 leads to rebalancing of Bcl-2 family members towards the pro-apoptotic BH3-only proteins in CLL. Furthermore, MLN4924 reversed stroma-mediated protection from fludarabine and the alkylators. In summary, we were able to abrogate the protective effect of the CLL microenvironment in vitro using MLN4924, a Nedd8-activating enzyme inhibitor. MLN4924 reduced NFκB activity in CLL cells, led to re-expression of the pro-apoptotic BH3-only proteins and interfered the chemokine network thus carrying a potential to disrupt cell homing. These results suggest a rationale for clinical investigation of MLN4924 as a novel tool to target microenvironment in CLL. 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: 2013
    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: Investigational New Drugs, Springer Science and Business Media LLC, Vol. 39, No. 4 ( 2021-08), p. 1099-1105
    Abstract: BI 836826 is a chimeric immunoglobulin G1 antibody targeting CD37, a transmembrane protein expressed on normal and malignant B cells. This open-label, phase Ib, dose-escalation study was conducted to determine the recommended phase II dose (RP2D) of BI 836826 + ibrutinib in patients with relapsed/refractory chronic lymphocytic leukemia (CLL). Eligible patients received 420 mg/day of ibrutinib with escalating doses of BI 836826. BI 836826 was administered in 4-week cycles. After Cycle 12, patients achieving complete response (CR), CR with incomplete marrow recovery, or minimal residual disease-negative partial response could continue to receive BI 836826 + ibrutinib every 4 weeks for ≤ 12 additional cycles. Patients received either 100 mg ( n  = 3) or 200 mg ( n  = 3) BI 836826 + ibrutinib. In the 100 mg BI 836826 cohort, one patient received two cycles and two patients received 22 cycles of BI 836826. In the 200 mg BI 836826 cohort, patients received 12, 16 and 20 cycles of BI 836826, respectively. All patients discontinued BI 836826 and continued ibrutinib outside the trial. No dose-limiting toxicities were reported in the maximum tolerated dose (MTD) evaluation period. As the trial was discontinued before the MTD was reached, the RP2D was not determined. Grade 3/4 adverse events (AEs) were predominantly hematological. Pseudomonal bacteremia was the only drug-related AE of special interest. BI 836826 + ibrutinib did not exceed the MTD at doses up to 200 mg in patients with CLL. However, RP2D and MTD were not formally established, as the sponsor discontinued the trial.
    Type of Medium: Online Resource
    ISSN: 0167-6997 , 1573-0646
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2009846-7
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 3482-3482
    Abstract: RNA splicing dysregulation is a hallmark of cancers and underlies the onset and progression of diseases. Chronic lymphocytic leukemia (CLL) is one of the most common adult leukemia in western countries. Spliceosome mutations occur in ~20% of patients. However, the mechanism for splicing defects in spliceosome unmutated CLL cases remains elusive. Through an integrative transcriptomic and proteomic analysis of primary CLL samples, we discovered proteins involved in RNA splicing are post-transcriptionally upregulated. Coupled with clinical annotation, we found spliceosome protein abundance is an independent risk factor and associated with poor prognosis. Splice variants found in CLL are highly overlapped with those driven by high spliceosome abundance but not splicing factor mutations, indicating high spliceosome abundance contributes to genetic lesion-independent splicing defects. To identify potential regulators for spliceosome, we proteome-widely analyzed the proteins that highly correlated with splicing factors expression. Analysis of 113 CLL samples has consistently identified METTL3 upregulation with positive correlation with 77.6% of detected splicing factors. METTL3 is an RNA methyltransferase that modifies N6-methyladenosine (m6A) on mRNA and regulates the translation of m6A-installed transcripts. m6A level on mRNA is increased in CLL cells with differential m6A highly enriched on splicing related transcripts. Moreover, high METTL3 expression in CLL is also associated with poor clinical outcomes. These results suggested that METTL3 translationally controls splicing factors through m6A and plays a role in CLL progression. Toward this end, we demonstrated that METTL3 is essential for CLL growth in both in vitro and in vivo studies. Knock out (KO) and pharmaceutical inhibition of METTL3 decreased cell growth and splicing factor expression. Overexpression of wildtype but not catalytic mutant METTL3 restored both defects in METTL3 KO cells, indicating that the regulation of splicing factor is m6A-dependent. To dissect the underlying mechanism, we performed an integrated Ribo-seq, RNA-seq, and MeRIP-seq on CLL cells with or without METTL3. KO of METTL3 decreased overall translation efficiency (TE) with RNA splicing as the most significantly affected pathway. Splicing factors with reduced TE displayed either hypo-m6A at stop codon region or hyper-m6A at CDS regions upon METTL3 KO, as direct or indirect targets of METTL3. Moreover, we found that m6A at stop codon and CDS regions regulates splicing factor translation via ribosome recycling and ribosome pausing, respectively. Taken together, our results uncovered a novel regulatory axis for METTL3 that controls splicing factor translation and contributes to CLL progression. Our study highlights a post-transcriptional layer of m6A modification as a major contributor to genetic lesion-independent splicing defects in CLL. Citation Format: Yiming Wu, Meiling Jin, Mike Fernandez, Kevyn Hart, Aijun Liao, Stacey M. Fernandes, Tinisha McDonald, Zhenhua Chen, Daniel Röth, Lucy Ghoda, Guido Marcucci, Markus Kalkum, Raju K. Pillai, Alexey V. Danilov, Jianjun Chen, Jennifer R. Brown, Steven T. Rosen, Tanya Siddiqi, Lili Wang. METTL3-mediated m6A modification controls splicing factor abundance and contributes to CLL progression [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3482.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    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...