Ihre E-Mail wurde erfolgreich gesendet. Bitte prüfen Sie Ihren Maileingang.

Leider ist ein Fehler beim E-Mail-Versand aufgetreten. Bitte versuchen Sie es erneut.

Vorgang fortführen?

Exportieren
  • 1
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 2526-2526
    Kurzfassung: BACKGROUND: ALL cells are physically anchored in the bone marrow (BM) microenvironment through a network of adhesion molecules. Adhesion also creates intracellular signals that regulate proliferation and cell death. We have previously identified integrin α4 (α4) as a critical molecule for such cell-adhesion-mediated drug resistance in pre-B ALL. In chronic lymphocytic leukemia (CLL), α4-mediated activation of the PI3K/AKT pathway was reported. In ALL, stromal cell protection of B-lineage ALL cells has also been shown to require active Akt. In addition, we have shown that treatment with CAL-101, a commercially available PI3Kδ inhibitor, de-adheres pre-B ALL cells from the α4-counter-receptor VCAM1, downregulates p-Akt and induces apoptosis in primary pre-B ALL cells. However, the microenvironmental stimuli that activate PI3K/AKT in ALL remain unknown. PI3Kδ can be activated by extracellular signals upon engagement with extracellular matrix, however specific ligands remain elusive, the identification of which would be critical to understand the mechanism of PI3Kδ inhibition. These studies were designed to determine which extracellular ligands activate PI3K/AKT in patient-derived (primary) pre-B ALL cells and whether CAL-101 affects adhesion and migration of ALL cells. METHODS: Four samples ofpatient-derived (primary) pre-B ALL cells, co-cultured with murine calvaria-derived mesenchymal stromal (OP9) cells were treated with a commercially available PI3Kδ inhibitor, CAL-101 (Selleckchem), or its vehicle DMSO control. Culture plates with and without transwells were used for in vitro assays. Annexin V/7-AAD staining was used for viability determination by flow cytometry and Western Blot was used for determination of total Akt and p-Akt expression. RESULTS: To determine which ligands activate p-Akt, ALL cells were adhered to fibronectin (FN), VCAM-1 (both counter-receptors of integrin α4), laminin-1 (hLam-1; counter-receptor for integrin α6) and albumin (BSA). OP9 cells were used in direct contact with ALL cells or separated through a transwell insert. α-MEM or RPMI were used as media controls. hVCAM-1 and hLam-1 activated p-Akt in one out of three samples. In all 3 samples, OP9 cells activated p-Akt compared to media control, but only in direct contact, not when separated through a transwell. This suggests that direct adhesion of ALL cells to stromal cells rather than diffusible factors are associated with p-Akt activation. Next, we determined if CAL-101 de-adheres ALL cells independently from the ligands ALL cells are bound to. Primary pre-B ALL cells were plated in triplicates onto FN, VCAM-1, hLam1-coated 96-well plates and incubated with CAL-101 (10µM) ± HUTS-21 antibody (20µg/mL). HUTS-21 is an antibody that binds only to the active form of integrin β1, CD29, and can activate outside-in signaling via integrin β1 stimulation. CAL-101 inhibited cell-adhesion from FN (15±4.2% vs. 51±9.9%, p=0.04) and VCAM-1 (54.6 x±8.6%vs. 95.8±3.8%, p=0.03), but not from hLam1 indicating that it acts specifically on the VCAM-1/FN-mediated adhesion of ALL cells. HUTS-21 restored cell-adhesion of ALL cells to FN (67.0±1.4% vs. 81±7.1%, p=0.19) and VCAM-1 (42.5±16.2% vs. 50.6±8.6%, p=0.59) indicating adhesion recovering upon HUTS-21 addition to CAL-101-treated cells. Next, we determined if CAL-101 has an impact on migration of ALL cells. ALL cells were treated with CAL-101 (10μM) or DMSO control for 24 hours and added to a migration assay. We observed that migration of ALL cells to stromal cell-derived factor 1alpha (SDF-1alpha; CXCL12) was significantly decreased (1.7 x103 ±0.3x103 vs. 3.1x103 ±2.4x103 p=0.004), while migration to OP9 cells was partially inhibited compared to DMSO control (2.1 x103 ±0.5x103 vs. 4.2 x103 ±0.5x103 p=0.005). CONCLUSION: Taken together, our data demonstrate that the PI3Kδ/AKT pathway is stimulated by extracellular matrices and that PI3Kδ inhibition with CAL-101 inhibits adhesion of ALL cells to FN/VCAM-1 but not Lam1, which can be restored using HUTS-21. Data derived from further studies will determine the potential of the FDA-approved PI3Kδinhibitor Idelalisib as a novel therapy for pre-B ALL. Disclosures Wayne: NIH: Patents & Royalties; Medimmune: Honoraria, Other: travel support, Research Funding; Kite Pharma: Honoraria, Other: travel support; Pfizer: Honoraria; Spectrum Pharmaceuticals: Honoraria, Other: travel support, Research Funding.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2015
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 4591-4591
    Kurzfassung: The liquid biopsy allows assessment of multiple biological analytes over time to provide dynamic temporal information with the potential for improving clinical management and guiding treatment decisions. While the promise of liquid biopsies for prediction and response monitoring are intensely investigated, the pre-analytic variables are of primary concern for its implementation in diagnostic clinical medicine, including such categories as collection method, shipping conditions, and sample storage. Here we utilize an integrated high-definition single cell analysis (HD-SCA) workflow for genomic analysis of rare cells and cfDNA from the liquid biopsy to characterize the effects of pre-analytical variation and reproducibility of data analysis from the same cohort of patients. The results presented here confirm consistent rare cell enumeration and morphometric characterization between 24h and 48h time to assay (TTA), with a high efficiency and capacity for both copy number variation (CNV) and single nucleotide variation (SNV) analysis at the single cell level. Additionally, the freezing process neither diminishes or increases the DNA quantity, nor does it affect the DNA quality for CNV or SNV analysis. The integration of genomic information is imperative to reveal essential disease-implicated mutational profiles, but effective molecular diagnostic tests require reproducible coverage over a broad dynamic range. The performance of the HD-SCA platform quantified here may be utilized as a guide for implementation into patient care and/or research biorepository processes. Citation Format: Stephanie Nicole Shishido, Mariam Rodriguez_Lee, Anand Kolatkar, Liya Xu, Sara Restrepo-Vassalli, Lisa Welter, Anders Carlsson, Emily Greenspan, Shelley Hwang, Kathryn Waitman, Jorge Nieva, Kelley Bethel, James Hicks, Peter Kuhn. Integration of genomic analysis and assessment of pre-analytic variables in the HD-SCA workflow: A technical validation study [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4591.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2018
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 3763-3763
    Kurzfassung: BACKGROUND: The bone marrow is known to shelter leukemia cells from chemotherapy and contributes to the survival of chemotherapy resistant residual cells, termed minimal residual disease (MRD). We have studied in situ the location of MRD+ ALL cells using a xenograft model of primary ALL cells and have found a novel co-localization of megakaryocytes (MK) with ALL cells. Mature hematopoietic cells have been implicated in modifying the local normal hematopoietic stem cell environment including MK. We hypothesize that MK are associated with the survival of MRD+ ALL cells. For this purpose, we tested the role of MK cells in maintenance and chemoprotection of ALL cells. METHODS: Patient-derived (primary) pre-B ALL cells were engrafted into non-irradiated NOD/SCID IL2Rγ-/- (NSG) mice. Leukemia-bearing mice received 4 weeks of chemotherapy treatment (Vincristine, Dexamethasone, L-Asparaginase; VDL). MRD status of mice was confirmed by detection of human CD45 + CD19+ leukemia cells in the bone marrow by flow cytometry. In situ location of the MRD+ ALL cells was determined by histological analysis and quantitation was performed by Fiji Image J. For in vitro studies, primary pre-B ALL cells were co-cultured for up to 2 days with murine calvaria-derived stromal OP9 cells or MK isolated from C57/BL6 BM primed for with murine thrombopoietin (mTPO) and sorted by flow cytometry for CD41+ MK. Annexin V/7-AAD staining was used for viability determination by flow cytometry. Boyden chamber system with either OP9 cells or MK cells seeded on the bottom and ALL cells on the top of the system was used for migration assays. RESULTS: In situ analysis of MRD+ ALL recipient mice showed that cells MRD+ ALL cells (huCD45+) are located in close proximity to MKs, with 11.57±2.94% of MRD+ ALL cells lying directly adjacent to MKs (0-5μm distance to MK). To further assess the role of MKs in ALL survival in vitro, we compared if MK cells can sustain proliferation and viability of primary leukemia cells like the OP stromal co-culture model that we have established previously. MKs were isolated from BM of C57BL/6 mice by FACS sorting for CD41+ cells from BM primed with mTPO. The sorted population showed a 90% purity of CD41+ cells. MKs were able to maintain ALL cell proliferation 1.90e6 cell count ± 0.48e6 cell count on day 2) and provide chemoprotection from VDL treatment (77.24 ± 2.03% on day 2), which was similar to the effect of OP9 cells on sustained proliferation and viability. Interestingly ALL cells cultured with MKs had a slight reduction in G2/M-phase (8.46±0.31%) 3 days after culture set up without treatment compared to cultures with OP9 stromal cells (13.59±0.14%; P-value 〈 0.0005). In a migration assay, MKs stimulated migration of ALL cells (5.42e5 ±0.72 migrated cells) significantly more than OP9 stromal cells (2.92e5 ±0.72 migrated cells) over a 24 hour period (P-value = 0.0132). Using the SDF1α inhibitor AMD3100 (100µM), migration of ALL cells was only partially inhibited (2.92e5 ±0.72), suggesting additional MK produced factors influence mobilization of human leukemia cells besides SDF1α. Using the recombinant forms of stromal cell-derived factor 1 (SDF1α) and von Willebrand factor (VWF), ALL cell migration was successfully stimulated over 24 hours (6.25e4 ±1.25 and 3.33e4 ±0.72 migrated cells, respectively), and this effect was inhibited using AMD3100, small molecule inhibitor of CXCR4, and anti-VWF antibody respectively. CONCLUSION: Here we show for the first time that co-culture of MK and primary pre-B ALL cells supports their proliferation, viability and protection from chemotherapy similar to murine OP9 stromal cells. Our data warrants further investigation of the underlying mechanism. Disclosures No relevant conflicts of interest to declare.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2015
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 4
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 3807-3807
    Kurzfassung: BACKGROUND: Acute myeloid leukemia (AML) is the most common acute leukemia in adults. Acute myeloid leukemia comprises approximately one-fifth of pediatric leukemias and is the seventh most common pediatric malignancy. In children, relapse following primary therapy approaches 40%, and the 5-year event-free survival (EFS) rate is only approximately 50%. Treatment is dominated by generic chemotherapeutic agents. Novel therapeutic strategies are highly warranted. The bone marrow microenvironment has been shown to promote cell adhesion-mediated drug resistance in leukemia cells. Breaking adhesive bonds of AML cells with their protective niche to mobilize them from the bone marrow to the peripheral blood may make drug treatment more efficient. Our studies have suggested the adhesion molecule CD49d as an anchor molecule for ALL and AML cells in the bone marrow. However, as of today, no drug targeting CD49d is approved for use in leukemia. Here, we evaluate a novel human specific CD49d targeting antisense, ATL1102, in clinical development for Multiple Sclerosis, in human AML cells. METHODS: We determined CD49d expression in the human AML cell line HL-60 treated with a CD49d targeting antisense ATL1102 and antisense control by qPCR and flow cytometry. Annexin V/DAPI and BrdU stainings were used for viability determination and cell cycle assay respectively by flow cytometry. A NOD/SCID IL2Rγ-/- (NSG)xenograft model of human HL-60 cell line was used for an in vivo mobilization assay. RESULTS: To assess the on-target effect of ATL1102 on CD49d, HL-60 cells were nucleoporated with either ATL1102 or control antisense.mRNA expression of CD49dwas significantly decreased by ATL1102 treatment cells (85.2%±15.4 expression inhibition using ATL1102 1µM after 24h compared to control, p 〈 0.001) as assessed by RT-PCR. The FACS analysis 72 hours after treatment revealed a significant decrease of surface expression of CD49d in a dose-dependent manner (99%±0.4 (1µM, *), 87.9%±8.7 (3µM) and 57.8%±7.2 ATL1102 (10µM, ***), 55.9±13.5 (30µM, **) vs 99.7%±0.1 for control antisense (30 µM), P 〈 0.001, n=3). No significant effect on apoptosis or cell cycle was observed after ATL1102 treatment. We also evaluated the in vivo effect of ATL-1102 on mobilization of leukemia cells in a pilot experiment. For this purpose, HL-60 cells (5x106/per mouse) were injected via the tail vein in sublethally irradiated NSG mice. Presence of human ALL cells (hCD45) was determined weekly by flow cytometry of white blood cells isolated from peripheral blood (PB). 23 Days post-leukemia injection, mice were treated with either antisense control (CTRL) (n=3), ATL1102 (50mg/kg, n=2). Peripheral blood was drawn before and 24 hours after ATL1102-treatment. ATL1102 induced a strong mobilization of AML cells to the PB of leukemia-recipient mice compared to control antisense treated-mice (69.1% and 87.7% vs 1.1%, 0.2% and 28.1% for ATL1102 (50mg/ml) and CTRL treated-mice respectively. The mobilized cells show a decrease of surface expression of CD49d (16.8%±9.2% vs 32.8%±16.7%), although this was not of statistical significance in this pilot experiment. Experiments to repeat this assay with large numbers of mice are in progress as well as experiments to determine the initial location of the mobilized AML cells and synergy of ATL1102 with chemotherapy are ongoing. CONCLUSION: We demonstrate that ATL1102 can efficiently decrease CD49d expression in AML cell line in vitro and in vivo, and that ATL1102 leads to mobilization of AML cells to the peripheral blood. Disclosures Wayne: NIH: Patents & Royalties; Medimmune: Honoraria, Other: travel support, Research Funding; Kite Pharma: Honoraria, Other: travel support; Pfizer: Honoraria; Spectrum Pharmaceuticals: Honoraria, Other: travel support, Research Funding. Tachas:Antisense Therapeutics Ltd: Employment, Equity Ownership, Patents & Royalties.
    Materialart: Online-Ressource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Society of Hematology
    Publikationsdatum: 2015
    ZDB Id: 1468538-3
    ZDB Id: 80069-7
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 4459-4459
    Kurzfassung: Metastatic breast cancer patients have a high risk of progression and face poor prognosis overall, with about one third (34%) surviving 5 years or more. In rare instances (2-4% of cases) patients with metastatic breast cancer have ERBB2 (HER2) activating mutations, but are HER2 negative (non-amplified. Neratinib is a potent, irreversible inhibitor that binds HER2 and inhibits downstream signaling. We used the previously validated high-definition single cell analysis (HD-SCA) workflow to investigate the clinical significance of circulating tumor cells (HD-CTCs) in HER2-mutant, HER2 non-amplified, post-menopausal metastatic breast cancer patients starting neratinib and fulvestrant combinational therapy. Genomic analysis of the cell-free DNA (cfDNA) and single cell HD-CTCs was conducted to monitor tumor evolution and identify potential mutational variants unique to the patient’s clinical response. A subset of 5 patients presented here were from the MutHER (NCT01670877) and/or SUMMIT (NCT01953926) trials. Patients had an average of 5.4 lines of therapy before enrollment, variable hormone receptor status, and ERBB2 mutations at diagnosis and during treatment. HD-CTC enumeration alone was not sufficient to predict clinical response. Interestingly, treatment pressure was shown to lead to an observable change in HD-CTC morphology and genomic instability, suggesting these parameters may inform prognosis. Single cell copy number variation (CNV) analysis indicated that the persistence or development of a clonal population of HD-CTCs during treatment was associated with a worse response. Hierarchical clustering analysis of the single cells across all patients and timepoints identified distinct aberrant regions shared among patients, comprised of 26 genes that are similarly affected and may be related to drug resistance. Additionally at the cfDNA level, we identified new mutations in ERBB2, PIK3CA, and TP53 that arose due to treatment pressure in a patient with poor response, further informing us on the dynamics of the cancer genome over the course of therapy. The data presented in this small cohort study demonstrates the feasibility of real-time molecular profiling of the cellular and acellular fractions of the liquid biopsy using the HD-SCA platform. Additional insight is warranted to determine the potential use of morphometric and genomic analysis as a prognostic tool to advance personalized oncology. Citation Format: Stephanie Nicole Shishido, Rahul Masson, Lisa Welter, Liya Xu, Anishka D'Souza, Darcy Spicer, James Hicks, Janice Lu, Peter Kuhn. Morphologic and genomic characterization of circulating tumor cells in patients with ERBB2 mutant HER2 non-amplified metastatic breast cancer treated with neratinib [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4459.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2019
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Bibliothek Standort Signatur Band/Heft/Jahr Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie auf den KOBV Seiten zum Datenschutz