ALBERT

All Library Books, journals and Electronic Records Telegrafenberg

feed icon rss

Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
Filter
Collection
  • 11
    Publication Date: 2015-12-03
    Description: Background: Clinical studies have shown that some pts with CML-CP who achieve deep, sustained molecular response (MR) on BCR-ABL1 tyrosine kinase inhibitor (TKI) therapy are able to stop treatment and maintain treatment-free remission (TFR). Because more pts achieve deep MR with NIL vs IM, a higher proportion of pts on NIL may be eligible to attempt TFR. Currently, 4 studies (ENESTgoal, ENESTop, ENESTfreedom, and ENESTpath) are evaluating TFR after NIL in pts with CML-CP. ENESTgoal is an open-label phase 2 study of TFR after second-line NIL in pts who achieved major MR (MMR; BCR-ABL1 ≤ 0.1% on the International Scale [IS]) but not MR4.5 (BCR-ABL1IS ≤ 0.0032%) on IM. The MMR to MR4.5 eligibility window was based on the assumption that approximately one-third of pts within this range of MR would achieve MR4.5 within 2 years of switching to NIL on study. Due to slow enrollment and a higher than expected screen failure rate (primarily due to pts not being in the specified MR window), the sample size was reduced to 59 pts, and the NIL consolidation phase was changed to 2 years (Figure). Methods: Adult (aged ≥ 18 years) pts with Philadelphia chromosome-positive CML-CP who achieved MMR but not MR4.5 (confirmed in a central laboratory) after ≥ 1 year of IM therapy were switched to NIL 300 mg twice daily (BID) upon enrollment. On study, pts who achieve MR4.5 within 2 years of switching to NIL and maintain deep MR during a subsequent 2-year NIL consolidation phase are then eligible to attempt TFR (ie, stop NIL). Approximately 20 pts are expected to become eligible to attempt TFR. Real-time quantitative polymerase chain reaction (RQ-PCR) monitoring is performed by a central laboratory every 3 months on study and more frequently during the TFR phase. During the TFR phase, pts with loss of MMR (per protocol amendment) are required to re-initiate NIL. The primary endpoint is the molecular relapse-free rate 6 months after attempting TFR. Herein we present an early analysis of pts who switched from IM to NIL in ENESTgoal. Results: Fifty-nine pts were enrolled by January 9, 2015 (median age, 54 years [range, 26-74 years]); 66% of pts were male, and80% were Caucasian. Baseline Sokal risk scores were as follows: high, 5 pts (9%); intermediate, 9 pts (15%); low, 32 pts (54%); unknown, 13 pts (22%). Median prior IM treatment duration was 64 months (range, 13-163 months). As of the data cutoff date (March 30, 2015), 49 pts (83%) were on study (monitoring phase, n = 32; consolidation phase, n = 16; TFR phase, n = 1), and 10 pts (17%) had discontinued (monitoring phase, n = 8; consolidation phase, n = 2). Reasons for study discontinuation included withdrawn consent (n = 4), adverse events (AE; n = 2 [grade 1 transient ischemic attack and grade 4 pericardial effusion]), unsatisfactory therapeutic effect (n = 2), administrative problems (n = 1), and abnormal laboratory values (n = 1). The median NIL treatment duration on study was 11.5 months (range, 2.7-18.5 months). AEs were reported in 56 pts (95%), the majority of which were low grade. Grade 3/4 AEs included elevated lipase (10%); rash (3%); and elevated amylase, hypophosphatemia, bronchospasm, headache, hyperglycemia, leukocytosis, non-cardiac-related chest pain, small-intestinal obstruction, squamous cell carcinoma, pericardial effusion, and vomiting (2% each). NIL-related AEs (≥ 5%; all-grade) included rash (27%); fatigue (14%); pruritus (12%); lipase increased (10%); abdominal pain and constipation (8% each); fatigue, headache, and palpitations (7% each); and abdominal discomfort, alopecia, nausea, and weight decreased (5% each). There were no QTcF values 〉 500 ms and no deaths. A total of 19 pts (32%) achieved confirmed MR4.5, and the median time to MR4.5 was 119 days (range, 56-448 days). In the consolidation phase, the median follow-up was 153 days (range, 11-434 days), and the median duration of MR4.5 was 97 days (range, 11-434 days). Per the original protocol, 1 pt entered the TFR phase after 1 year of consolidation and had BCR-ABLIS levels of 0.0241% at 60 days and 0.0216% at 90 days after attempting TFR, triggering re-initiation of NIL. Conclusion: After switching from IM to NIL, 32% of pts achieved confirmed MR4.5 with a median treatment duration of 11.5 months. Safety results are consistent with previously reported NIL studies. Results from longer-term follow-up in ENESTgoal and those from other ongoing studies will provide a better understanding of the role of NIL in enabling pts to achieve TFR. Figure 1. Figure 1. Disclosures Ritchie: Incyte: Speakers Bureau; Celgene: Speakers Bureau. Deininger:Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Gilead: Research Funding; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Incyte: Consultancy, Membership on an entity's Board of Directors or advisory committees; Ariad: Consultancy, Membership on an entity's Board of Directors or advisory committees; Bristol-Myers Squibb: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding. Erba:Novartis: Consultancy, Speakers Bureau; Novartis: Consultancy, Speakers Bureau; Incyte: Consultancy, Speakers Bureau; Seattle Genetics: Consultancy, Research Funding; Amgen: Consultancy, Research Funding; Incyte: Consultancy, Speakers Bureau; Amgen: Consultancy, Research Funding; Seattle Genetics: Consultancy, Research Funding; Celgene: Consultancy, Speakers Bureau; Celgene: Consultancy, Speakers Bureau; Millennium/Takeda: Research Funding; Millennium/Takeda: Research Funding; Celator: Research Funding; Celator: Research Funding; Astellas: Research Funding; Sunesis: Consultancy; Astellas: Research Funding; Pfizer: Consultancy; Sunesis: Consultancy; Daiichi Sankyo: Consultancy; Pfizer: Consultancy; Ariad: Consultancy; Daiichi Sankyo: Consultancy; GlycoMimetics: Other: Data Safety and Monitoring Committees; Ariad: Consultancy; Jannsen (J&J): Other: Data Safety and Monitoring Committees ; GlycoMimetics: Other: Data Safety and Monitoring Committees; Jannsen (J&J): Other: Data Safety and Monitoring Committees. Savona:Celgene: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Karyopharm: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Incyte: Membership on an entity's Board of Directors or advisory committees, Research Funding. Warsi:Novartis Pharmaceutical Corporation: Employment. Paley:Novartis Oncology: Employment. Dautaj:Novartis Pharmaceutical Corporation: Employment. Lin:Novartis: Employment. Mauro:Ariad: Consultancy; Bristol-Myers Squibb: Consultancy; Novartis: Consultancy, Research Funding; Pfizer: Consultancy.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 12
    Publication Date: 2016-12-02
    Description: Background:Spleen tyrosine kinase (SYK) is a nonreceptor cytoplasmic tyrosine kinase primarily expressed in cells of hematopoietic lineage. Constitutive activation of SYK in acute myeloid leukemia (AML) has been reported and targeted inhibition of SYK induced differentiation in vitro and demonstrated anti-leukemia activity in AML mouse models. SYK has also been shown to directly phosphorylate the FLT3 receptor, modulating its activation and possibly promoting its role in leukemogenesis. Entospletinib is an orally bioavailable, selective inhibitor of SYK shown to be clinically active in B-cell malignancies. Here we evaluate the combination of entospletinib in patients with untreated AML using a 14-day window phase to assess single-agent activity, then adding standard intensive chemotherapy. Methods: In this phase 1b/2 study (NCT02343939), patients age 18 to 70 years with previously untreated AML, preserved organ function, and ECOG ≤ 2 were eligible to receive dose escalated entospletinib for 14 days as monotherapy (days -14 to 0) followed by combination with daunorubicin 60 mg/m2/d, cycle 1 day 1 to 3, and cytarabine 100 mg/m2/d, cycle 1 day 1 to 7. All patients received entospletinib monotherapy for up to 14 days prior to starting induction. Chemotherapy could be initiated after 5 days of monotherapy (and entospletinib continued for 4+ weeks) in patients with leukemia-related complications necessitating chemotherapy. Patients enrolled to dose level (DL) 0 and DL 1 received entospletinib 200 mg po BID and 400 mg po BID, respectively. Patients with residual disease two weeks after chemotherapy received a second induction cycle identical to the first. Entospletinib was continued without interruption until remission was assessed at count recovery. Results:Twelve patients enrolled with a median age of 54 (range, 18-69) years. Patients were in the following European LeukemiaNet genetic risk groups: favorable (n=1), intermediate I (n=3), intermediate II (n=2), and adverse (n=4), respectively. Three patients were not evaluable for dose limiting toxicity (DLT) assessment and were replaced (due to detection of CNS disease requiring non-study therapy (n=1), and withdrawal of consent unrelated to drug toxicity (n=2)). Single-agent entospletinib during the window period was well tolerated; toxicities after combination with intensive chemotherapy were common and typical. Among 3 patients treated at 200mg BID, no DLT was observed. Of 3 patients treated at 400mg BID, a patient with documented fungal pneumonia developed grade 3 pneumonitis that was possibly related to entospletinib. Although this did not meet DLT criteria, DL 1 was expanded with 3 additional patients, none of whom experienced DLT. Overall, the most common non hematologic adverse events (inclusive of intensive chemotherapy periods) were febrile neutropenia, nausea, and diarrhea. Based on this clinical experience and compiled pharmacokinetic data demonstrating lack of benefit to further dose escalation, 400 mg BID was selected as the recommended phase 2 dose. Responses were seen at both levels. Among the 3 patients treated at 200 mg BID, two required a second induction but all achieved a complete remission (CR) (3/3; 100%). Of the 6 patients treated at 400mg BID, none required a second induction and the CR rate was also 100%. Remarkably, an 18 year old male with 11q23-rearranged AML achieved morphologic and cytogenetic CR after only the 14 day entospletinib monotherapy window (prior to chemotherapy). Another patient with 11q23-rearranged AML had significant platelet response during the window period (this patient refused disease evaluation by marrow aspiration prior to chemotherapy). Conclusions: Entospletinib appears to have significant clinical activity in AML, and its combination at doses up to 400mg BID with intensive chemotherapy is well tolerated. An extended phase 2 program is now underway. Patients with 11q23-rearranged AML may be uniquely sensitive to SYK inhibition by entospletinib. Detailed molecular analysis of these patients is ongoing and will be presented. Disclosures Walker: Gilead Sciences: Research Funding. Bhatnagar:Karyopharm: Research Funding. Marcondes:Gilead Sciences: Employment, Equity Ownership. DiPaolo:Gilead Sciences: Employment, Equity Ownership. Abella-Dominicis:Gilead Sciences: Employment, Equity Ownership.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 13
    Publication Date: 2016-12-02
    Description: Background: Treatment-free remission (TFR; ie, stopping tyrosine kinase inhibitor [TKI] therapy without loss of response) has been demonstrated in multiple trials in patients (pts) with chronic myeloid leukemia in chronic phase (CML-CP) with stable deep molecular response (DMR) after long-term TKI therapy. Enabling TFR requires frequent molecular response (MR) assessments near the limit of detection (LOD) for real-time quantitative polymerase chain reaction (RQ-PCR) before and after stopping treatment. Digital PCR (dPCR) may have the potential for more sensitive detection of BCR-ABL1. ENESTgoal is an open-label phase 2 study in pts receiving imatinib (IM) who achieved major MR (MMR; BCR-ABL1 ≤ 0.1% on the International Scale [IS]) but not MR4.5 (BCR-ABL1IS ≤ 0.0032%) and were switched to nilotinib (NIL) upon enrollment. Methods:Pts (aged ≥ 18 years) with Philadelphia chromosome-positive CML-CP who had MMR but not MR4.5 after ≥ 1 year of IM were switched to NIL 300 mg twice daily in a monitoring phase of up to 2 years. Pts who achieved confirmed MR4.5 during the monitoring phase entered a 2-year NIL consolidation phase (changed via protocol amendment from randomization to 1 or 2 years of consolidation). Pts with no confirmed loss of MR4 (BCR-ABL1IS ≤ 0.01%) during consolidation were eligible to stop NIL and enter the TFR phase; pts who were randomized to a 1-year consolidation and had already entered TFR prior to the protocol amendment continued in the TFR phase. Pts with molecular relapse, defined as confirmed loss of MMR (2 samples within ≈ 4 weeks) during TFR, reinitiated NIL. RQ-PCR was performed every 3 months before TFR, monthly for the first 6 months of TFR, and every 2 months thereafter during TFR. When sufficient sample was available, dPCR was also performed for samples from the consolidation and TFR phases that were expected to be below the RQ-PCR LOD (BCR-ABL1IS 〈 0.0032%). Results:As of the data cutoff (May 9, 2016), 59 pts were enrolled (median follow-up in monitoring phase, 297 days): median age, 54 years; male, 66%; median prior IM duration, 64 months. Of these,42 pts (71%) remained on study (monitoring phase, n = 9; consolidation phase, n = 29; TFR phase, n = 1; reinitiation phase, n = 3). Median NIL exposure on study was 21 months (range, 〈 1-31 months). A total of 39 pts (66%) achieved confirmed MR4.5 (median follow-up in consolidation phase, 336 days); median time to MR4.5 was 220 days (range, 56-757 days). As of the data cutoff, 25 pts did not have confirmed loss of MR4 during consolidation, and 4 pts had entered the TFR phase; of these 4, 3 had only 1 year of NIL consolidation. Three pts restarted NIL after 70, 99, and 153 days in the TFR phase (final BCR-ABL1IS during TFR: 0.5722%, 0.0216%, and 0.2258%, respectively); all regained MR4.5 with NIL retreatment. As of the data cutoff, 1 pt remained in TFR (duration, 138 days; BCR-ABL1 undetectable at last measurement). However, longer follow-up is needed to determine the rate and duration of TFR. Adverse events (AEs) reported in ≥ 10 pts during NIL treatment included fatigue (n = 22; 37%), constipation (n = 15; 25%), rash (n = 14; 24%), headache (n = 12; 20%), abdominal pain and pruritus (n = 11; 19% each), and diarrhea, lipase increased, and weight decreased (n = 10; 17% each). The majority of events were grade 1/2. Serious NIL-related AEs were unstable angina, arterial stenosis, pericardial effusion, peripheral arterial occlusive disease, and transient ischemic attack (n = 1 each). Seven pts discontinued from the study due to AE/abnormal laboratory value; the remaining study discontinuations were due to withdrawal of consent (n = 6) and unsatisfactory therapeutic effect (n = 4). No deaths occurred on study. In 97 samples from pts in the consolidation and TFR phases that had undetectable BCR-ABL1 by RQ-PCR, dPCR analysis was performed. BCR-ABL1 was detected by dPCR in 11 of these samples (11%); none of these pts have relapsed. Conclusion: The majority (66%) of pts with MMR but not MR4.5 on IM achieved confirmed MR4.5 after switching to NIL on study. The safety profile of NIL was generally consistent with that seen in previous NIL studies. dPCR detected BCR-ABL1 transcripts in samples from some pts with undetectable BCR-ABL1 by RQ-PCR. Achievement of sustained DMR and maintenance of TFR in ENESTgoal continue to be evaluated with ongoing follow-up; future dPCR analysis will compare BCR-ABL1 transcript levels at the time of stopping NIL in pts who did or did not have molecular relapse. Disclosures Ritchie: Incyte: Speakers Bureau; Celgene: Speakers Bureau; Arian: Speakers Bureau; Novartis: Honoraria; Pfizer: Honoraria. Pinilla-Ibarz:Novartis: Consultancy; Abbvie: Consultancy, Speakers Bureau; Gilead: Consultancy, Speakers Bureau; Janssen: Consultancy, Honoraria; Pharmacyclics: Consultancy, Speakers Bureau. Deininger:Gilead: Research Funding; BMS: Consultancy, Research Funding; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Pfizer: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Incyte: Consultancy, Membership on an entity's Board of Directors or advisory committees; CTI BioPharma Corp.: Membership on an entity's Board of Directors or advisory committees; Celgene: Research Funding; Bristol Myers Squibb: Consultancy, Research Funding; Ariad: Consultancy, Membership on an entity's Board of Directors or advisory committees. Erba:Incyte: Consultancy, DSMB, Speakers Bureau; Novartis: Consultancy, Speakers Bureau; Juno: Research Funding; Pfizer: Consultancy; Astellas: Research Funding; Amgen: Consultancy, Research Funding; Celgene: Consultancy, Speakers Bureau; Jannsen: Consultancy, Research Funding; Gylcomimetics: Other: DSMB; Daiichi Sankyo: Consultancy; Seattle Genetics: Consultancy, Research Funding; Agios: Research Funding; Millennium Pharmaceuticals, Inc.: Research Funding; Celator: Research Funding; Ariad: Consultancy; Sunesis: Consultancy. Radich:BMS: Consultancy; Novartis: Consultancy, Research Funding; Ariad: Consultancy. Savona:Takeda: Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees; Gilead: Membership on an entity's Board of Directors or advisory committees; Sunesis: Research Funding; TG Therapeutics: Research Funding; Amgen Inc.: Membership on an entity's Board of Directors or advisory committees; Ariad: Membership on an entity's Board of Directors or advisory committees; Incyte: Membership on an entity's Board of Directors or advisory committees, Research Funding. Dautaj:Novartis: Employment. Wolff:Novartis: Employment. Mauro:BMS: Consultancy; Novartis: Consultancy; Ariad: Consultancy; Pfizer: Consultancy.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 14
    Publication Date: 2016-12-02
    Description: Acute myeloid leukemia (AML) is an incurable disease with 5 year survival rates of 10% in patients over 60 years. Poor tolerance to chemotherapy, chemo resistance and high rate of relapse warrants less toxic and more effective regimens in AML. OSU-2S is a novel non-immunosuppressive derivative of FTY720, a sphingosine analogue. The promising in-vitro and in-vivo activity of OSU-2S against a number of leukemias and lymphomas, and other malignancies such as hepatocellular carcinoma, impelled us to evaluate the activity of OSU-2S in AML. The potent cytotoxic activity of the OSU-2S (5µM, 24hrs) in AML cell lines HL-60, MV411 and MOLM13 (n=3; HL-60: p=0.008; MV411: p=0.04; MOLM13: p=0.0094) encouraged us to evaluate the effect of OSU-2S in primary leukemic cells from AML patients. OSU-2S (5µM, 24 and 48 hrs) demonstrated significant cytotoxic activity against AML cells, including high risk FLT3-ITD mutated AMLs, (n=13, p
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 15
    Publication Date: 2016-12-02
    Description: Background: Patients undergoing allogeneic hematopoietic stem cell transplant with reduced-intensity conditioning (RIC-alloSCT) rely primarily on the graft-versus-tumor (GVT) effect to prevent relapse. Although relapse remains high in this setting, previous data suggest modifications of allograft composition could produce enhanced GVT effect and improve outcomes. Prior retrospective studies suggested higher total nucleated cell (TNC) and CD8 dose correlate with improved overall survival (OS) and a reduction in relapse among all patients undergoing RIC-alloSCT. The aim of our study was to further investigate this association by comparing transplant outcomes with detailed graft immunophenotyping data including T-cell and NK-cell activation and maturation status and CD34 cell doses in patients with AML and MDS undergoing RIC-alloSCT. Methods: We performed a retrospective analysis using data from consecutive patients with AML and MDS who underwent RIC-alloSCT at a single center from 2010-2015 who had graft immunophenotyping data available. We compared transplant outcomes with TNC per kilogram (kg) and dose of CD3, CD4, CD8, CD34, and NK cells along with selected activation and maturation subsets. A competing risk regression analysis was conducted to examine the association between cell dose/kg and risk of relapse and graft versus host disease (GVHD). Overall survival and relapse free survival (RFS) were assessed utilizing the Cox proportional hazard model. Estimates of survival probability were determined by the Kaplan-Meier survival function. The log-rank test was used to compare OS and RFS among patients. Univariable and multivariable regression analyses were performed. Disease Risk Index (DRI) was used to stratify risk among patients. To identify optimal cutoffs (OC) of continuous variables for a specific outcome, a Classification and Regression Tree (CART) algorithm was utilized. Results: Our study included 142 patients who underwent RIC-alloHSCT for AML (n=97, 68%) and MDS (n=45, 32%). The vast majority of patients (98%) received fludarabine and busulfan conditioning with 65% receiving anti-thymocyte globulin. All patients received tacrolimus-based GVHD prophylaxis. Overall relapse rate was 37% at 3 years. Overall survival was 49% with median follow-up of 3.2 years. Total Nucleated Cell Dose: In multivariable analysis controlling for disease, comorbidity index, and performance status, a high infused TNC dose (10.6 - 16.8 x 108 cells/kg) correlated with improved overall survival (HR 0.27, 95% CI [0.12 - 0.59], p=0.001) and relapse free survival (HR 0.31, 95% CI [0.15 - 0.64], p=0.002). However, a very high TNC dose (OC 〉16.8 x 108 cells/kg) correlated with significantly worse OS (HR 2.84, 95% CI [1.16 - 6.98], p=0.023) and trended toward worse RFS (HR 2.09, 95% CI [0.88 - 4.93], p=0.093). Higher TNC doses were also correlated with an increased risk of chronic GVHD (HR 2.16, 95% CI [1.28 - 3.67], p=0.004, OC 9.8 x 108 cells/kg). Cell Subsets: Very low CD3 cell doses (OC
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 16
    Publication Date: 2006-11-16
    Description: Histone acetylation plays a key role in the regulation of gene expression. Histone hyperacetylation is associated with chromatin opening and gene transcription, while histone hypoacetylation is associated with chromatin condensation and gene silencing. Abnormal histone hypoacetylation mediated by aberrant activity of histone deacetylases (HDACs) has been found to be associated with silencing of tumor suppressor and growth inhibitory genes in malignant cells. HDAC inhibitors (HDACIs) can relieve HDAC-mediated gene silencing and thereby induce normal patterns of cell cycle, differentiation and apoptosis in malignant cells. HDACI OSU 42 is a novel hydroxamate tethered phenylbutyrate derivative that was designed and synthesized at our institution, and exhibited IC50s at submicromolar level, compared with millimolar level for other members of this classes of HDACIs such as valproic acid (VPA). We characterized the activity of this compound in acute myeloid leukemia (AML) cells. It is known that the fusion proteins AML1/ETO and PML / RAR alpha that characterized t(8;21) and t(15;17) AML silence target genes through recruitment of HDACs to their promoter regions. Therefore we utilized AML1/ETO-positive Kasumi-1 and PML/RARA-positive NB4 cells to test the activity of HDACI OSU 42 and used THP-1 cells, characterized by AF9/MLL fusion gene, as a control. We hypothesized that by virtue of the fusion genes, Kasumi-1 and NB4 are more susceptible to HDACI treatment. IC50s for proliferation inhibition in Kasumi-1 cells treated with HDACI OSU42 were 71.8±14.3nM for 24hr and 31.3± 0.4nM for 48hr, significantly lower than VPA (2.0mM for 24hr, 0.9mM for 48hr). The IC50s for NB4 were 237.7±6.5nM for 24hr and 119±6.4nM for 48hr. As a contrast, IC50 for THP-1 was 507.3±68.3nM for 48hr. HDACI OSU42 inhibited 80% of total HDAC activity at 125nM in both Kasumi-1 and NB4; 30nM HDACI OSU42 induced hyperacetylation of histone H3 and H4. Apoptosis analysis showed that nearly 60% more of Kasumi-1 and NB4 underwent apoptosis after treated with 1μM of HDACI OSU42 for 24hr, compared with their untreated control. On the other hand, the same treatment only induced 15% more of THP-1 undergoing apoptosis. Apoptotic effect of HDACI OSU42 was mediated by activation of caspase 9 and caspase 3. Cell cycle analysis demonstrated that treatment of Kasumi-1 and NB4 with 150nM of HDACI OSU 42 inhibited cell cycle progression and arrested 20% to 30% more cells at S phase or G2/M phase, whereas this treatment had not effect on cell cycle progression of THP-1. This was consistent with the up-regulated expression of p21 at both transcription level and protein level. Q-PCR data suggested that Kasumi-1 and NB4 treated with HDACI OSU42 expressed ~10 folds of p21 higher than untreated cells. Chromatin immunoprecipitation assay revealed 10 to 50 folds increase in acetylation level of histone H3 and H4 associated with p21 promoter. Kasumi-1 and NB4 cells also show differentiation ability (increase in CD14 and CD 13 expression by flow cytometry) when treated with 30nM of HDACI OSU42, whereas THP-1 remained undifferentiated. These results support the activity of HDACI OSU42 as a new potent HDACI in AML.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 17
    Publication Date: 2006-11-16
    Description: Patients with acute myeloid leukemia (AML) who receive therapy for their disease are at significant risk for infections. One risk factor for infections is hyperglycemia which can often be observed in medically ill patients. In post-operative critically ill patients, intensive insulin therapy has been shown to reduce the incidence of nosocomial infections. (Van den Berghe, 2001) In one study of patients with acute lymphoblastic leukemia, hyperglycemia during induction therapy was associated with more infections and shorter disease free survival. (Weiser, 2004) We hypothesized that hyperglycemia is associated with worse outcomes in AML due to increased incidence and severity of infectious complications. We performed a retrospective cohort study to determine if there was an association between hyperglycemia and hospital mortality in hospitalized patients with AML. Four-hundred and sixty-four adult patients were identified through a search of our administrative databases as having the diagnosis of acute myeloid leukemia over a three year period. These individuals experienced 1,004 admissions to the hospital. The occurrence of increasing hyperglycemia, defined as the proportion of glucose assessments above normal (〉110 mg/dL), during all hospital days was associated with increased odds of hospital mortality [OR 1.24, 95% CI 1.13–1.36, p
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 18
    Publication Date: 2014-12-06
    Description: Introduction: Donor T cells have been known to be the most powerful effectors for acute graft-versus-host-disease(aGVHD). The HMG-CoA reductase inhibitors, statins, have been shown to inhibit T-helper 1(Th-1) driven responses, reduce pro-inflammatory cytokines while increasing the migration of anti-inflammatory cytokines T-regs and T-helper-2 cells(Th-2), with a potential to reduce aGVHD. We analyzed the effect of atorvastatin on the immune reconstitution pattern on patients enrolled in a phase II study evaluating the efficacy of atorvastatin as GVHD prophylaxis in patients undergoing HLA-matched-related donor AHSCT in which donors and patients were exposed to atorvastatin. The results of this study were compared to historical matched controls at Ohio State where neither related donor nor patients were exposed to any cholesterol lowering medications. Methods: Forty patients (statin gp) were enrolled (NCT01491958) between March 2012 and January 2014. Atorvastatin at 40mg/day orally was administered to sibling donors, starting 14-28 days before the anticipated 1st day of stem cell collection. In AHSCT recipients, atorvastatin (40mg/day) was administered from day -14 to day +180 (or until stopping immunosuppression, toxicity, development of grade [Gr] II-IV aGVHD, or severe chronic GVHD (cGVHD). In addition all recipients received standard GVHD prophylaxis with tacrolimus and methotrexate. Historic matched controls were 25 patients (and related donors) not on any cholesterol lowering medications (non-statin gp) and had mobilized peripheral blood (PB) allograft samples (day 0) and PB samples on days 30 and 100 post AHSCT collected. We analyzed the absolute numbers of T, NK, NKT, B cells and immunophenotypic characterization of their activation status. Results: Median duration of atorvastatin prophylaxis in statin donors was 14 days (range 13-26) and in statin patients 132 days (32-400). Baseline patient’s characteristics were well matched between the groups with respect to age, sex, donor/recipient sex, intensity of regimen, disease and disease status at transplant and number of stem cells infused. The clinical results of this study are presented in a separate abstract in which we found no statistically significant difference in clinical outcomes including aGVHD and cGVHD between the two gps. While there were no differences in the B and total T cells reconstitution in all time points, the statin gp allografts contained less NK and T-regulatory cells (T-regs) than the non-statin gp (Table 1). This reduction of the NK and T-regs, appeared to affect the reconstitution of these subsets post-transplant as reflected by their continued decrease on days 30 and 100 post-transplant compared to the non-statin gp (Table 1). Conclusion: while we found no differences in clinical outcome with the addition of atorvastatin, it did result in a statistically significant decrease in NK and T-reg cells for the statin gp in the graft when administered to donors, and on days 30 and 100 post HSCT in the recipients. We are yet to determine the significance of this. Table Statin gp vs. non-statin gp allograft for % markers Median (range)Statin N=40 Median (range)Non-statin N=25 Wilcoxon test p-value CD3-/CD5616+ 8.00 (2.30-26.90) 10.75 (4.7-20.0) 0.0374 CD3+/CD134+ 1.40 (0.30-8.70) 3.1 (0.5-11.2) 0.0367 CD3+ / CD86+ 0.20 (0.05-0.56) 0.3 (0.1-0.5) 0.0148 CD4+/CD25+/CD127- 0.10 (0-1.60) 1.0 (0-4.7)
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 19
    Publication Date: 2014-12-06
    Description: INTRODUCTION: Acute graft-versus-host disease (aGVHD) remains a significant cause of morbidity and mortality following allogeneic stem cell transplantation (allo-SCT). CD74, also known as the HLA class II invariant chain, is expressed on the surface of antigen presenting cells (APCs) and involved in pro-survival signaling. Milatuzumab is a humanized IgG1K monoclonal antibody that interacts with CD74 leading to rapid internalization and cytotoxicity independent of antibody crosslinking. Milatuzumab, therefore, has potential to inhibit APC-mediated T-cell proliferation and decrease the risk of aGVHD. In a xenogeneic SCID mouse model, milatuzumab administered prior to SCT prevented the development of GVHD, suppressed circulating cytokines, and reduced the mortality of the SCID mice (Chen, BBMT, 2013). METHODS: We are performing an open-label, single-center phase 1 dose escalation trial to determine the maximum tolerated dose of milatuzumab when added to standard GVHD prophylaxis in the setting of reduced-intensity conditioning (RIC) for HLA matched (≥8/8) sibling or unrelated donor SCT. Patients are premedicated with dexamethasone and receive milatuzumab intravenously on Days -7, -4, -1, and +7. The initial starting dose was 8 mg/kg with continued dose escalation to 16 mg/kg and 20 mg/kg in cohorts of 3-6 until unacceptable toxicity. Dose limiting toxicities are defined as graft failure, any ≥ grade 4 organ toxicities, or ≥ grade 4 infusion reactions attributable to milatuzumab (CTCAE v4.0). Expected hematologic toxicities from SCT were excluded from this definition. Acute and chronic GVHD was recorded according to Consensus and NIH criteria, respectively. RESULTS: Seven patients have been treated on trial with median follow up of 53 days post SCT (average 105 days; range 13-234). The median age is 59 years (range 26-70). Patients had a variety of hematologic malignancies (3 AML, 1 MDS, 1 ALL, 1 HD, & 1 MCL). Median Sorror comorbidity score (CMI) was 1 (range 0-8). Patients were in complete remission prior to SCT except for one MCL patient with partial response to prior therapy. Recipients received RIC with fludarabine (30 mg/m2 days -7 to -3) and busulfan (0.8 mg/kg Q6 hours days -4 & -3 x8 doses) along with standard GVHD prophylaxis including tacrolimus and methotrexate (D+ 1, 3, 6, & 11). Donors were 8/8 HLA-matched siblings (UPNs 2, 4, & 7) or matched unrelated (UPNs 1, 3, 5, & 6). All patients received four doses of milatuzumab. Three individuals were initially treated with 8 mg/kg (DL1) and 3 received 16 mg/kg (DL2). At DL2, two of 3 patients had grade 5 sepsis with one having grade 4 respiratory failure. As a result, three more patients will be enrolled to DL1 to further assess safety; one has been treated thus far. The most common toxicities attributable to milatuzumab were grade 1-2 including GI complaints, nasal congestion, fever, parathesia, and urticaria. One patient had grade 1-2 infusion reactions on two occasions. Four of 7 patients had cutaneous aGVHD with 2 of 4 experiencing grade 1, one with grade 2, and one with grade 4 (table 1). One patient experienced grade 1 GI aGVHD. The grade 4 aGVHD (UPN 6) was histologically and phenotypically indistinguishable from toxic epidermal necrolysis/Stevens-Johnson syndrome and that patient was treated with brentuximab for steroid-refractory cutaneous GVHD. He subsequently died due to polymicrobial sepsis. Two other patients died, one from relapsed AML (UPN 4) and one from staphylococcal aureus sepsis (UPN 5). CONCLUSIONS: While the grade 5 events in DL2 (16 mg/kg) were not considered specifically attributable to milatuzumab, DL1 has been expanded to further evaluate safety of this dose. Four patients have been enrolled at DL1 with only grade 1 and 2 aGVHD reported, either cutaneous or upper GI. We will continue to accrue at DL1 and pending tolerability may amend the protocol to test an intermediate dose of 12 mg/kg. Milatuzumab remains an intriguing potential agent to prevent aGVHD. Abstract 1168. Table 1: Matched RIC allo-SCT recipients treated prophylactically with milatuzumab for prevention of aGVHD UPN Age Disease CMI Dose Level aGVHD Follow up 1 59 AML 4 1 D+20 (grade 2/stage 3 cutaneous) Alive D+234 2 70 AML 0 1 D+22 (grade 1/stage 2 GI & cutaneous) Alive D+181 3 26 HD 1 1 D+35 (grade 1/stage 1 cutaneous) Alive D+186 4 51 AML 1 2 none Relapse D+26 (Death D+53) 5 66 MDS 8 2 none Death D+13 6 57 ALL 0 2 D+16 (grade 1/stage 2); D+34 (grade 4/stage 4) cutaneous Death D+48 7 59 NHL (MCL) 0 1 none Alive D+22 Disclosures Goldenberg: immunomedics: Employment. Wegener:immunomedics: Employment.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
  • 20
    Publication Date: 2006-11-16
    Description: Background: Tandutinib is an orally bioavailable small molecule inhibitor of FLT3, c-KIT, and PDGFR with a single-agent MTD of 525 mg b.i.d. Tandutinib demonstrated single agent anti-leukemic activity in patients with relapsed/refractory AML with FLT3 ITD mutations, with ≥50% decreases in bone marrow and peripheral blast counts in 12/25 patients and 1 CR without platelet normalization. Since tandutinib is synergistic with cytarabine and daunorubicin in vitro, we sought to determine the MTD of tandutinib in combination with standard induction chemotherapy in patients with newly diagnosed AML, with or without FLT3 ITD mutations. Methods: A starting dose of Tandutinib 200 mg b.i.d was administered during induction and consolidation therapy, and for an additional 6 months. Induction therapy consists of cytarabine 200mg/m2/day IVCI, days 1–7, plus daunorubicin 60mg/m2/day, days 1–3. Consolidation therapy is given as 2–4 cycles of standard (3000mg/m2 IV every 12h, days 1, 3, 5) or in older patients modified (2000mg/m2/day IV, days 1–5) high-dose cytarabine. DLT is defined as failure to recover marrow function (ANC ≥500/μL; platelets ≥20,000/μL), or grade 3/4 non-hematologic toxicity not resolved to grade 2 (except anorexia, alopecia, fatigue) by day 42 of induction therapy, or any unexpected grade 3/4 non-hematologic toxicities. Results: 29 patients have been enrolled: median age 60y (range 26–83); 13M, 16F; 23 de novo, 6 secondary AML; 9 with unfavorable cytogenetics; 5 with FLT3 ITD mutations. Cohort 1 consisted of 7 patients treated with continuous daily dosing of tandutinib 200 mg b.i.d. Due to GI intolerance, the protocol was amended so that tandutinib was administered only on days 1–14 of induction therapy and each cycle of consolidation. Under the amended schedule 8 patients were treated with tandutinib 200 mg b.i.d. (Cohort 2) and 14 patients have been treated with tandutinib 500 mg b.i.d. (Cohort 3). Full safety and efficacy data are available for the 15 patients in cohorts 1 and 2. Diarrhea, nausea and vomiting have been the most common drug-related AEs, and were more frequent with continuous daily dosing of tandutinib. GI tolerance in Cohort 2 has been acceptable, with no patients requiring termination or reduction in tandutinib for GI toxicity. Although continuous dosing was not feasible, no DLTs were seen in Cohorts 1 or 2; one DLT consisting of obtundation not clearly related to tandutinib during induction occurred in Cohort 3, One patient in Cohort 3 experienced non-dose limiting generalized muscle weakness, which reversed within 24 hours after discontinuation of tandutinib. Tandutinib was restarted at a reduced dose in this patient without recurrence. 5/7 patients in Cohort 1 and 6/8 patients in Cohort 2 achieved a CR. PK data have been collected for all 15 patients in Cohorts 1 and 2; median steady state tandutinib concentration was 195 ng/mL (range: 52–486). Conclusions: Tandutinib 200 and 500 mg b.i.d. in combination with standard therapy for newly diagnosed AML appears well tolerated using the amended dosing schedule (days 1–14). Updated results from Cohort 3 (tandutinib 500 mg b.i.d) will be presented.
    Print ISSN: 0006-4971
    Electronic ISSN: 1528-0020
    Topics: Biology , Medicine
    Location Call Number Expected Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...