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
  • American Society of Hematology  (10)
  • 1
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 3969-3969
    Abstract: Background AFM11 is a bispecific, tetravalent T cell-engaging antibody construct binding to CD19 on B cell origin malignant cells such as B-precursor acute lymphoblastic leukemia (B-ALL) and to CD3 on T cells. By engaging CD3-positive T cells, AFM11 elicits T cell-mediated killing of CD19-positive (CD19+) leukemia and lymphoma cells. In vivo anti-tumor activity of AFM11 was investigated in a Raji tumor xenograft model in non-obese diabetic/severe combined immunodeficiency (NOD/scid) mice reconstituted with human peripheral blood mononuclear cells (PBMC). Tumor growth in all AFM11-treated animal groups was significantly reduced. In the highest dose group, all animals achieved complete tumor remission. (Reusch et al., 2015). An ongoing Phase 1 study assesses safety, tolerability, pharmacokinetics (PK), pharmacodynamics (PD) and preliminary efficacy of AFM11 in patients with relapsed/refractory (R/R) CD19+ B-ALL. Methods Patients (pts) with relapsed or refractory CD19+ B-ALL are being enrolled into escalating cohorts of 1-6 pts. The primary objective of the study is to determine the maximum tolerated dose (MTD) of AFM11 administered as a 2-week continuous intravenous (CIV) infusion. AFM11 is administered over the first 2 weeks (wks) of each 4 wk cycle for up to 3 cycles. Pts with rapidly progressing disease receive pre-treatment with 200 mg cyclophosphamide and 10 mg/m2 dexamethasone over 3-5 days before initiating AFM11. A lower starting dose is employed during the first wk of cycle 1 and escalated to a target dose during the second wk of cycle 1 and all subsequent cycles. An accelerated titration design is used until toxicity is observed, followed by a classical 3+3 design. PD activity is assessed by flow cytometry of peripheral blood lymphocytes and serum cytokine measurements. Tumor response is evaluated by local bone marrow and peripheral blood laboratory results between days 15 and 18 of each cycle. Results As of June 29, 2018, fourteen pts (8 female/6 male) have been treated in 5 cohorts. The median age is 41.5 years (range 19-67) and the median number of prior therapies is 5 (range 1-12). AFM11 was well-tolerated with no dose-limiting toxicities (DLTs) observed in the first 5 cohorts. The study switched to 3+3 design in cohort 3 due to the occurrence of grade 2 AFM11-related events in cohort 2. Enrollment into cohort 6 is ongoing and the MTD has not yet been reached. The most frequent (≥2 pts) AFM11-related adverse events were pyrexia (29%), myalgia (14%), and tremor (14%). Most of the events were Common Terminology Criteria for Adverse Events (CTCAE) v4.03 grade 1-2, with one grade 3 and no grade 4 events observed. Transient and reversible neurological events occurred in 3 of 14 (21%) pts: grade 1 paresthesia (n=1), grade 1/grade 2 tremor (n=1 each), grade 2 lethargy (n=1); and grade 3 altered state of consciousness (n=1). The grade 3 event occurred during the third cycle of treatment and was managed with treatment interruption and steroids and resolved within 48 hours. The pt then completed the third cycle at a reduced AFM11 dose without incident. Peripheral B cell reductions were observed in multiple pts in cohorts 4 and 5 and notable cytokine release was detected in two pts. Two pts achieved complete remission with complete hematological recovery (CR): one pt in cohort 4 achieved CR after the first cycle and progressed 2 wks later; one pt in cohort 5 achieved CR after the first cycle which was sustained after the second and third cycles and was assessed as minimal residual disease (MRD) negative after cycle 3. As a result, this pt became eligible to receive stem cell transplantation upon study completion. Both CR pts had peripheral B-cell aplasia after the first few days of treatment. Updated data will be presented. Conclusions The CD19/CD3-targeting tetravalent bispecific T cell engager AFM11 was well-tolerated in pts with R/R B-ALL across the first 5 cohorts of an ongoing Phase 1 study, and the MTD has not been reached. Pyrexia was the most frequently observed related adverse event. Transient neurological events were observed and were consistent with those associated with other CD19-targeted therapies. Peripheral B cell reductions and complete remissions observed in pts treated in the higher dose cohorts suggest that AFM11 is active in pts with R/R B-ALL and that the study is nearing determination of the therapeutic dose and schedule. Disclosures Salogub: Affimed: Research Funding. Mayer:Novartis: Research Funding; Roche: Research Funding; Eisai: Research Funding; Johnson & Johnson: Research Funding; Affimed: Research Funding. Folber:Affimed: Research Funding. Grosicki:Affimed: Research Funding. Skotnicki:Affimed: Research Funding. Prochwicz:Affimed: Research Funding. Myasnikov:Affimed: Research Funding. Gural:Affimed: Research Funding. Schoenborn-Kellenberger:Affimed: Consultancy. Brindley:Affimed: Consultancy. Knackmuss:Affimed: Employment. Schwarz:Affimed: Employment. Schmich:Affimed: Employment. Choe-Juliak:Affimed: Employment. Strassz:Affimed: Employment. Alland:Affimed: Employment. Doubek:AbbVie: Consultancy, Research Funding; Gilead: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria; Roche: Consultancy, Honoraria; Affimed: Research Funding; Novartis: Consultancy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 104, No. 11 ( 2004-11-16), p. 4071-4071
    Abstract: Platelet anti-aggregants play a major role in the treatment of cardiovascular disorders. Recently it has been suggested that a significant proportion of patients fail to respond to these agents according to different testing methods. We have developed a new method for evaluating the response to platelet anti-aggregants, the modified Cone and Plate(let) Analyzer (CPA) test. The method is based on the observation that pre-activation of platelets by an agonist will result in reduced platelet deposition on a polystyrene surface under arterial flow conditions (the phenomenon of platelet adhesion refractoriness). In vitro studies demonstrated that the basic platelet adhesion (surface coverage, SC 12.3±6.8%) was significantly reduced in response to pre-incubation of the sample (for 1 min) with arachidonic acid (AA, SC 2.1±1.5%), adenine diphosphate (ADP, SC 1.3±0.6%) and epinephrine (EPI, 2.9±0.9%). This effect was selectively inhibited by aspirin (for the response to AA, SC 8.1±3.8%), and by 2-Methylthio-AMP triethylammonium (2-MeSAMP), a selective inhibitor of P2Y12 (for the response to ADP, SC 4.8±2.0%), p & lt;0.001. The potential application of this method for monitoring the effect of aspirin therapy (daily doses of 100, 300 and 500 mg), among 20 healthy volunteers was investigated in comparison to turbidimetric aggregometer. All volunteers responded to aspirin in the three doses according to the aggregometer test (reduced max. aggregation from 88±8% to 12.4±7%), with no evidence for a dose response manner in this dose range. Basic platelet adhesion as tested by the CPA (SC 9.8±2.2%), was unaffected by aspirin in the three tested doses. However, the response to pre-incubation with AA was significantly inhibited by aspirin in all patients (SC of 0.6±0.3% before aspirin therapy and 3.5±1.3%, 4.4±1.7%, and 4.1±2.0% following therapy with 100, 200 and 500 mg aspirin per day, respectively, p & lt;0.001 for all doses), again with no dose response effect. This effect was specific for AA, since no difference in SC was observed when ADP was used as an agonist. A significant correlation between the effect of aspirin as tested by the modified CPA and by the turbidimetric aggregometer using AA as an agonist (R2=0.55) was observed. In conclusion, the CPA was found useful point-of-care method for testing the response of platelets to aggregating agonists as well as for monitoring the effect of anti-platelet drugs.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 136, No. Supplement 1 ( 2020-11-5), p. 48-49
    Abstract: Introduction: Primary central nervous lymphoma (PCNSL) is a rare type of non-Hodgkin lymphoma that may involve the brain, spinal cord, leptomeninges and vitreous. The median age at diagnosis is in the fifth decade and most patients (pts) present with significant neurologic deficit and a low performance status. The optimal treatment of PCNSL is controversial. High dose methotrexate (HDMTX) is a standard treatment of PCNSL and is more effective when given in combination with other CNS-penetrating medications, which, however, add to the toxicity of the treatment. We aimed to evaluate the effectiveness and the safety of the combination of rituximab, HDMTX, procarbazine and lomustine (R-MPL) in pts with PCNSL. Patients and methods: We retrospectively reviewed the files of PCNSL pts treated in Hadassah Medical Center, Jerusalem, Israel, between the years 2006-2019. The medical records were reviewed for demographic details and initial disease characteristics (age, sex, performance status, laboratory results, cerebrospinal fluid (CSF) content and tumor location), and for therapeutic details including chemotherapy protocol, toxicity, response to treatment and survival. We excluded pts who could not receive HDMTX. The R-MPL is a 42-day cycle protocol, consisting IV Rituximab 375 mg/m2 on days 1, 15 and 28, IV HDMTX 5 g/m2 (3.5 g/m2 for pts & gt; 60 Years (y)) on days 2, 15 and 29, PO procarbazine 100 mg/m2 (60 mg/m2 for pts & gt; 60 y) on days 3-9, PO lomustine 60 mg/m2 (40 mg/m2 for pts & gt; 60 y) on day 2. Six to nine intrathecal (IT) injections of MTX / cytarabine were included for pts with positive CSF cytology, tumor adjacent to ventricles, or per physician's decision. Rituximab was given for no more than 8 doses in total. A total of 3-4 courses of R-MPL were given. Responsive pts could proceed to autologous stem cell transplant (ASCT) with TECAM conditioning or 2 cycles of intermediate dose cytarabine (IDAC, 1.5 g/m2), 2 doses in each cycle. Those who achieved less than CR or had significant toxicity to R-MPL received additional ifosfamide/etoposide or high dose cytarabine or temazolamide or topotecan. Radiation was given only for salvage. Results: Fifty-two pts were included in the study. Median age was 62 years (range 28-94). Three (6%) had leptomeningeal involvement, thirteen (25%) had vitreoretinal involvement, 30 (58%) had involvement of the deep brain. Mean ECOG, IELSG and MSKCC scores were 1.98, 2.53 and 1.94 respectively. The median number of HDMTX doses was 8 (range 3-16). Forty-five (87%) pts completed at least one 42-day cycle of R-MPL. Among this group, 29 (64%) pts received at least 3 IT injections, 12 (27%) underwent ASCT, 10 (22%) received IDAC, and 7 (16%) received other chemotherapies. The median follow-up was 27 months (m) (range 0.4-140m). The overall response rate (ORR) was 79% (41/52), and the complete response (CR) rate was 52% (27/52). The median progression free survival (PFS) was 15.5m and the median overall survival (OS) was 27m. Among the 45 pts who received at least one R-MPL cycle, the ORR was 87% (39/45), the CR rate was 60% (27/45), the median PFS was 84m and the median OS was not reached (figure 1). Known prognostic factors correlated with OS: age (p & lt;0.01), ECOG performance status (p & lt;0.01), CSF protein (p=0.01) and IELSG/MSKCC scores (p=0.05/0.04). Patients who received at least 3 IT injections had a trend to longer PFS (32.4m Vs 19.2m, p=0.09), and a significant OS benefit (48.6m Vs 20.8m, p & lt;0.01). In a multivariable model, the effect of IT injections was independent of age, sex and IELSG score, and attenuated by number of HDMTX doses. ASCT/IDAC did not improve PFS/OS, however numbers were small. Achieving CR at the end of treatment strongly correlated with PFS/OS, but timing of best response (after one 42-day cycle Vs later) did not. Grade 3-4 adverse events included infections (21%) and kidney injury (13%). Two pts died during therapy, both in the 1st cycle due to disease progression. Ten pts (19%) discontinued therapy due to toxicity. No treatment related mortality (TRM) was documented. Conclusions: The R-MPL protocol achieved a favorable ORR/PFS/OS as described, with reasonable grade 3-4 toxicity and no TRM. The advantage of ASCT/IDAC could not be assessed due to small number of pts. IT injections (at least 3) and achievement of CR at the end of treatment correlated with survival. The R-MPL protocol was reported in previous trials only among the elderly, and should further be evaluated in all age groups. Disclosures Goldschmidt: Abbvie Inc: Consultancy, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2020
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 134, No. Supplement_1 ( 2019-11-13), p. 5091-5091
    Abstract: Introduction: On November 2018 the FDA approved the treatment of venetoclax together with hypomethylating agent (HMA) or low dose cytarabine (LDAC) for newly diagnosed AML patients who cannot tolerate intensive chemotherapy. There is limited data regarding the efficacy and safety of venetoclax in the setting of relapsed/refractory patients who received intensive chemotherapy for their disease. Methods: In this study, data were collected retrospectively from Israeli medical centers where venetoclax was used as an advanced line of treatment after intensive chemotherapy. Venetoclax was given as a combination with HMA or LDAC or as monotherapy. Results: 40 patients with AML were included in the study, 60% were Males. The median age at time of diagnosis was 67 years (range, 21-82). 27 (67.5%) patients had de-novo AML and 13 (32.5%) had secondary disease; 7 (17.5%) patients had a history of MDS and 6 (15%) of MPN. 37% of the patients had normal karyotype (20% of them had FLT3-ITD mutation), 23% had complex karyotype. Based on the 2017 ELN risk classification, 12.5% had favorable risk, 42.5% - intermediate and 45% had high risk. At presentation, 87.5% of the patients received the '7+3' protocol as induction therapy but only 45% achieved a CR. Ultimately, 57.5% of the patients achieved a CR post consolidation/ salvage therapy. 42.5% underwent allogeneic hematopoietic cell transplant (HCT); 71% received myeloablative conditioning and the others reduced intensity. The median time from diagnosis to starting venetoclax was 6 (1-64) months. Patients received venetoclax after a median of 2 (range, 1-4) prior lines of treatment (not including HCT). The median age at starting venetoclax was 68.5 years (range, 22-83). The ECOG performance status at starting venetoclax was 0-1 in 67.5% of the patients, 2 in 22.5% and 3 in 10% of the patients. The median daily dose of venetoclax was 400 mg (range: 100-800 mg). Venetoclax was given together with HMA (mostly azacitidine) in 62.5% of the patients and with LDAC in 22.5%. In 12.5% of the patients it was given as monotherapy and in 1 patient it was given with the FLAG-IDA regimen. The median number of cycles of venetoclax that were given so far is 2.75 (range: 0.5-14). None of the patients experienced tumor lysis syndrome (TLS). 27.5% of the patients did not survive more than 2 months. Of the 29 patients who survived longer than 2 months, 76% achieved neutrophil recovery and 77% of them recovered also their platelet count. In 52% of those who survived more than 2 months, CR/CRi was confirmed by bone marrow (BM) examination. Taken together, 76% of the patients who survived more than 2 months achieved blood cell count recovery and in 68% of them it was confirmed as CR/CRi by BM examination. At a median follow-up of 5.5 months, 35% of the patients are still alive and 57% of them are still on venetoclax. The leading cause of death (81%) was disease progression. The median OS from starting venetoclax of all the patients and of those who survived more than 2 months was 5.5 and 6.5 months, respectively (Figure 1). Conclusions: Venetoclax was given to high risk AML patients with relapsed/refractory disease after intensive chemotherapy and in around 40% of the patients also after HCT. The treatment was tolerable and no patient developed TLS. Among those who survived more than 2 months, the CR/CRi rate, or at least blood cell count recovery rate, was high (76%) and the median OS was 6.5 months. These retrospective data, albeit in a heterogeneous group of previously treated patients with AML, demonstrate that the combination of venetoclax with HMA or LDAC is safe and active also in AML patients with advanced disease. Its precise role for patients who relapse after intensive chemotherapy needs to be evaluated in a prospective clinical trial. Figure 1 Disclosures Wolach: Novartis: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Speaker; AbbVie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Other: Speaker. Rowe:BioSight: Consultancy. OffLabel Disclosure: Venetoclax for relapsed/ refractory AML
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2019
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 3487-3487
    Abstract: Abstract 3487 Background: Drug metabolism and disposition genes have a substantial impact on the pharmacology of many medications. For CML, the human organic cation transporter (hOCT1) actively transports imatinib mesylate (IM) into cells, thus is a crucial factor in the response to IM therapy, without a known effect on plasma levels. A number of single nucleotide polymorphisms (SNPs) are believed to affect the activity of hOCT1 and therefore may influence IM accumulation in cells. We therefore studied hOCT1 SNPs and correlated with IM plasma levels and clinical response to IM. We studied a SNP in exon 2 of hOCT1 (480C 〉 G, Phe160Leu, rs683369), which, in the homozygous state (GG), has been associated with treatment failure on IM (DH Kim, Clin Cancer Res., 15:4750, 2009), and a SNP in exon 7 (1222A 〉 G, Met408Val) which has been hypothesized as having increased activity (A Giannoudis, Haematologica 96(s2):87, 2011). Both of these are prevalent SNPs; the exon 2 SNP is found in 22% of Caucasians and the exon 7 SNP, in 60% of Caucasians (R Kerb, Cancer Letters 234:4, 2006). Methods: We studied 48 patients with CML in chronic phase (M:F 21:27 aged 17–89 years. Time since CML diagnosis was between 10 months and 18 years. IM therapy was between 10–128 months' duration. Most of the patients were on a dose of 400 mg/once daily, with the exception of 11 patients who were on doses 〉 400 mg/once daily (mean 609.1 mg/day +/− 70.1 mg), and 9 patients, whose dose was 〈 400 mg/once daily (mean 281 mg/day +/− 35.4 mg). Trough IM levels were performed by Novartis Pharmaceuticals on samples taken 24 hours after last dose, reported in ng/ml. Compliance was verified by a patient diary during the trough drug testing period, but not throughout the entire period of IM therapy. Genotyping for hOCT1 SNPs was performed on 36 patients. The exon 2 SNP was detected by PCR amplification and Dde I restriction enzyme analysis. The exon 7 was detected by PCR and restriction with BmgB1. Detailed molecular data was available on 36 patients, and partial molecular data on 8 others. Complete molecular remission (CMR) was defined as PCR negativity using RT-PCR until 2004, and thereafter, using a highly sensitive real time PCR assay. Results: The mean IM trough level for all 48 patients was 1,153.0 (+/−493.1). There was much variation between IM levels in individual patients and levels also varied by dose. Patients who were on 400 mg/day had a mean IM level of 1064.1 (+/− 399.2). Patients on higher IM doses ( 〉 400 mg) had IM levels averaging 1397.2 (+/− 493.1). Patients on doses 〈 400 mg/day had mean IM levels of 1057.3 (+/− 667.0), which was nearly identical to that of patients on 400 mg/day. Genotyping for the hOCT1 exon 2 SNP demonstrated that 66.7% of the 36 patients analyzed were homozygous for the wild type allele, 13.9% were heterozygous for the polymorphism and 19.4% were homozygous polymorphic. GG homozygotes had higher IM levels than CG/CC genotypes (1562.75 +/− 560 compared to 1310.6 +/− 470). This is because the patients were on doses greater than 400 mg/day (mean 483 mg/day compared to 405 mg/day). These patients took an average of 8.8 months to achieve a molecular CR and one transiently lost molecular CMR. Genotyping for the hOCT1 exon 7 SNP demonstrated that 47% of the 36 patients analyzed were heterozygous AG for the polymorphism and 53% of the patients were GG homozygotes. IM levels for the GG homozygotes were higher than for AG heterozygotes (1413 +/− 549.6 compared to 1254.0 +/− 393.9 ng/ml), despite the fact that they were on a slightly lower average dose (mean: 390.1 mg/day compared to 436.7 mg/day). For 13 of these GG homozygous patients, detailed molecular followup was available. Two of these patients never achieved CMR and 2 transiently lost CMR. The time to achieve complete molecular remission was generally long, 17.6 months (range 3–83 months). Importantly, 12 out of 13 of these patients with unsatisfactory clinical responses to IM were CC or CG at the exon 2 SNP, which are considered favorable genotypes for IM response (DH Kim, Clin Cancer Res., 15:4750, 2009). Conclusions: These data may suggest that the GG genotype for exon 7 in hOCT1 (rs628031), independent of the exon 2 (rs683369) genotype, is an adverse prognostic parameter despite adequate IM levels. These patients may be candidates for an alternate tyrosine kinase inhibitor. Further studies are necessary on larger groups of patients to confirm these results, which are important in view of the high frequency of the polymorphic rs628031 allele. Disclosures: Cohen: Novartis Pharmaceutical Company: Research Funding. Rund:Novartis Pharmaceutical Company: Honoraria, Research Funding.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2011
    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. 108, No. 11 ( 2006-11-16), p. 2550-2550
    Abstract: Although the transcriptional product of the (8;21) chromosomal translocation, the AML1-ETO protein, has numerous well established effects on the behavior of human hematopoietic cells, the specific cellular pathways perturbed by it are only partly known. Recently AML1-ETO has been shown to bind E-proteins (members of the class I basic helix-loop-helix family) via its NHR1 (nervy homology region one) domain, thus causing a replacement of the co-activator p300/CBP complex by the co-repressor hystone deacytelase complexes and abrogating the E-protein induced transcriptional activation (Zhang et al, Science 2004). The removal of amino acids 93 to 189 from the NHR1 domain (the ΔNHR1 mutant form) has completely reversed this inhibitory effect, and thus we have studied the biological effects of this deletion. To assess the difference in self-renewal capacity between cells transduced with the full length and the ΔNHR1 forms of AML1-ETO, CD34+ cells were plated into methylcellulose culture medium. On days 14 and 28 myeloid and erythroid colonies were scored, and the cells harvested and re-plated. The scoring on day 14 showed no significant difference in either the overall number or the type of colonies, with both the full-length AML1-ETO and the ΔNHR1 mutant showing a decreased erythroid colony formation, thus indicating a preserved negative effect on hematopoietic cell differentiation. However, on days 28 and 42 cells expressing the full-length AML1-ETO scored a significantly higher number of colonies than cells expressing the ΔNHR1 mutant, whereas cells transduced with the empty MIGR1 vector were unable to form any colonies after second re-plating. To assess the difference in growth potential, we have plated cells containing each of the constructs in IMD medium supplemented with 20% BIT and cytokines (SCF, FLT-3L, IL-6, TPO). Following one week in culture the cells were harvested, counted and then serially re-plated for five weeks. We observed a significantly higher number of cells expressing the full-length AML1-ETO as compared to the ΔNHR1 mutant at each weekly time point. Cells growing in the liquid culture were also weekly re-plated into methylcellulose culture medium. In this assay the loss of NHR1 domain significantly diminished the ability of AML1-ETO to maintain colony-forming progenitors in culture. To assess the difference in transcriptional activity of the different constructs, we have measured the expression of p21, and several other potential AML1-ETO target genes, by Real Time PCR. The expression of p21 was increased 14-fold by the full-length AML1-ETO, but only 2-fold by the ΔNHR1 mutant. We conclude that partial deletion of the NHR1 lowers the self-renewal capacity of transduced hematopoietic cells, decreases the proliferative advantage conferred by AML1-ETO, undermines its ability to maintain colony-forming units, and affects the transcriptional activation of AML1-ETO target genes. As the loss of NHR1 abolishes the E-protein induced transcriptional activation without affecting the binding of AML1-ETO to DNA, our findings identify an additional mechanism of action for AML1-ETO, independent of its dominant-negative effect on AML1 target genes.
    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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 1340-1340
    Abstract: Abstract 1340 Background: Drug metabolism/disposition genes have substantial impact on drug pharmacology. Single nucleotide polymorphisms (SNPs) may affect gene activity. For CML, the human organic cation transporter (hOCT1) transports imatinib mesylate (IM) into cells. The multidrug resistance gene MDR1 effluxes IM. MDR1 SNPs are correlated with response to IM (Dulucq et al, 2008). We studied hOCT1 and MDR1 SNPs and correlated them with plasma levels and clinical response to IM. We studied an exon 2 hOCT1 SNP (480C 〉 G, Phe160Leu) for which homozygosity (GG) is associated with IM failure (Kim, 2009), and an exon 7 SNP (1222A 〉 G, Met408Val) which correlated with major molecular remission (MMR) (Takahashi, 2010). Both are prevalent in Caucasians (exon 2 SNP: 22%, exon 7 SNP: 60%) (Kerb, 2006). Methods: We studied 84 chronic phase CML patients (pts) aged 17–89 years, followed from 16 months (mos) to 20 yrs from diagnosis (mean 94 mos). 19% were diagnosed during or prior to 2000. Time to initiating IM ranged from 〈 1 to 108 mos (median 〈 1 mo). IM therapy duration ranged 12–142 mos (mean 75). 61 pts took 400 mg/day, 14 pts took 〉 400 mg (mean 621 mg), and 9 pts took 〈 400 mg (mean 283 mg). Trough IM levels were performed on 81 pts by Novartis Pharmaceuticals. Treatment response was defined as per European Leukemia Net criteria. Until 2004, MMR was defined as PCR negativity using RT-PCR, and thereafter, using real time PCR (BCR/Abl 〈 0.1% on an international scale). Results: The overall mean IM trough level was 1,168 ng/ml (+/–477), and varied by dose. Pts on 400 mg had a mean level of 1145 (+/– 468), pts on 〉 400 mg had 1392 (+/– 472), and pts on 〈 400 mg had 909 (+/– 425). Table 1 shows genotype frequencies. hOCT1 exon 2 GG homozygotes had higher IM levels than CG/CC genotypes (1231 +/− 644 versus 1162 +/−463). hOCT1 GG patients were on slightly higher doses (mean 457 +/− 162 mg compared to 422 +/− 96 mg for CC/CG pts). hOCT1 exon 7 AA homozygotes had the same IM levels as did GG/AG pts (1178 +/− 477 for AA, 1177 +/− 476 for GG/AG). This is despite the fact that the AA homozygotes were on higher doses (500 +/− 134 mg for AA, 413 +/− 93 mg for GG/AG). Notably, 20% (2/10) AA patients failed IM compared to 11% (8/73) GG/AG pts. MDR1 2677 TT had lower than average IM levels (964 +/− 557) on a mean dose of 414 +/− 90 mg. Pts with genotypes 2677GG/GT had IM levels of 1195+/−473 on a slightly higher dose of 428 +/− 104 mg. MDR1 3435 TT homozygotes had similar IM levels to CC/CT pts (TT: 1127 +/− 434; CC/CT: 1188 +/− 434). TT patients were on slightly lower doses (404 +/− 77 compared to 434 +/− 111 mg). 82/84 of the pts are still alive and 82% are still on IM. 15 pts discontinued IM (10 treatment failures, 5 due to intolerance). Only two pts developed accelerated phase or blast crisis. Time to MMoR is seen in Table 2, as related to genotype. 20 pts lost molecular remission (LMR) at some point. Some of these had received IM as second line therapy (average 17.7 mos from diagnosis to starting IM). Half of the LMR events occurred when no second like TKIs were available so IM dose was increased. Half the pts with LMR switched to a newer TKI. Of treatment failures, there was a tendency to more A alleles in hOCT1 Exon 7 (AG or AA) and more T alleles in both MDR1 SNPs (GT/TT for 2677 and CT/TT for 3435). Conclusions: 1. IM levels are associated with clinical response in CML. Treatment failures may be caused by suboptimal IM dosing. Alternate TKIs may overcome this problem. 2. Failures were more likely in pts for whom IM was not front line therapy. 3. Genotypes seem to correlate with IM levels and with treatment response for both SNPs. The hOCT1 AA genotype was associated with a higher dose requirement and more treatment failures. TT genotype for both MDR1 SNPs correlated with longer time to MMR (Table 2). Disclosures: Rund: Novartis Corporation: Honoraria, Research Funding.
    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 ...
  • 8
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 3954-3954
    Abstract: Introduction: Progression-free (PFS) and overall survival (OS) rates for Primary mediastinal large B-cell lymphoma (PMBL) have risen to 84% and 92%, respectively, with the addition of rituximab to standard CHOP. Despite general acceptance of RCHOP as standard of care for Diffuse Large B-Cell lymphoma (DLBCL), many centers recommended alternative regimens for PMBL such as RMACOPB, RVACOPB, RCHOP-RICE and DA-EPOCH-R. The latter was adopted with great worldwide enthusiasm, despite its lack of proven superiority in randomized controlled trials (RCT). The benefits of DA-EPOCH-R include the omission of consolidation radiotherapy (RT), an attractive option in PMBL patients (pts), given their demographic profile-mainly females in their 3rd decade. We aimed to evaluate the PFS, the OS, the number of hospitalization days for treatment, and complications and the need for consolidation RT in a single center in the Rituximab era; where since 2007 over 80% of our pts were treated with the RCHOP-RICE regimen consisting of 4 courses of RCHOP followed by 3 courses of RICE. Methods: We reviewed the files of all PMBL pts who received 1st-line treatment in Hadassah Medical Center between 8/2002-10/2014, extracting clinical, laboratory and imaging data. Results: Of the 47 pts, 24 were treated with RCHOP-RICE (80% since 2007), 12 with RMACOPB, 3 with RVACOPB, 6 with RCHOP and 2 with DA-EPOCH-R. Pts were mainly female, with Stage I-II disease, and a high LDH level. Pt characteristics were comparable between the protocols (Table). In total, 21 (45%) of our pts received RT; only 3 pts (12%) treated with RCHOP-RICE compared to 18 pts (78%) treated with other protocols (p 〈 0.01). A mean of 11+8 hospitalization days/pt were needed to administer the RCHOP-RICE regimen, significantly more than required for other treatments combined (p 〈 0.01), except DA-EPOCH-R where the mean hospitalization days to administer 6 courses-=37+2 /pt (2 patients). Treatment-related toxicities did not differ between the groups. Late toxicity included advanced breast cancer in one pt who received RMACOPB and radiotherapy. The Deauville 5-point scale at interim was available for 39 pts, of whom 43% had an uptake 〈 /= to mediastinal blood pool, 33% had an uptake greater than the mediastinum and 〈 /= liver uptake and 23% had 〉 liver uptake. At the end of therapy the numbers were 68%, 23% and 9% respectively, for 35 pts who were evaluated. The median 5-year PFS and OS were 93% and 98% respectively, with no difference between treatment regimens. Conclusion: The RCHOP-RICE regimen does not appear inferior to other regimens, allows to omit RT in PMBL and demonstrated no significant late toxicities. Published phase 2 data on DA-EPOCH-R (93% EFS and 95% OS) do not demonstrate an advantage compared to the simpler regimens described here. RCTs are required to establish the standard for efficacy, efficiency and safety of care in PMBL. Table 1. Characteristics RCHOP-RICE Others All patients Number of patients 24 23 47 Median age 34 34 34 Female n(%) 16 (67) 15 (65) 31 (66) Stage 1-2 n(%) 17 (71) 21 (91) 38 (81) Median tumor size (cm) 10.2 10.5 10.2 High LDH n(%) 21 (87) 17 (81) 38 (84) Effusion n(%) 10 (42) 9 (39) 19 (40) Median 5 year PFS 90% 95% 93% Median 5 year OS 100% 95% 98% Disclosures Lavie: Pfizer: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees.
    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 ...
  • 9
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 1588-1588
    Abstract: Abstract 1588 Transcription factors and histones are similarly modified through acetylation, phosphorylation, ubiquitination and methylation, which impact on the transcriptional regulation of gene expression and various biological processes in normal and malignant hematopoiesis. The t(8;21) associated AML1-ETO fusion protein is found in 40% of the FAB M2 subtype of acute myeloid leukemia, but how the post-translational modification of AML1-ETO affects its leukemogenicity is largely unknown. Here we show that AML1-ETO directly interacts with the lysine acetyltransferase, p300, via the region containing NHR1 domain and that p300 can acetylate two lysine residues in AML1-ETO and AML1-ETO (exon 9a) in human and mouse leukemia cells. To understand the biological effects of AML1-ETO acetylation, we used human CD34+ cord blood cells as a preleukemia model. The maintenance of CD34+ cells by the acetylation defective form of AML1-ETO was 5 fold less than with AML1-ETO (p 〈 0.01) in the liquid culture assay, and unlike the effect of AML1-ETO, the number of the cobble stone area forming cells (CAFC) was not increased by the mutant AML1-ETO in CAFC assay. However, the block in erythroid and myeloid differentiation conferred by AML1-ETO was still seen in the AML1-ETO acetylation mutant transduced human CD34+ cells. We then approved the impact of acetylation on leukemogenicity using the AML1-ETO9a (AE9a) mouse leukemia model. Mice receiving AE9a acetylation mutant transduced fetal liver cells have not developed leukemia by Day 250, whereas all the mice receiving AE9a transduced cells died due to leukemia before Day 160, with a mean survival time of 109 days (p 〈 0.001). These results suggest that the acetylation of AML1-ETO is required not only for its self-renewal promoting effects and but also for the development of acute leukemia. To gain insight into the mechanisms of AML1-ETO acetylation, we performed luciferase assays and found that the AML1-ETO acetylation mutant lost the ability to activate an M-CSFR promoter driven reporter construct. Furthermore, the expression levels of AML1-ETO activated target genes related to self-renewal were not upregulated in AML1-ETO acetylation mutant transduced human CD34+ cells. These results indicated that the acetylation is crucial to AML1-ETO induced transcription activation. We have also been studying the role of the region containing NHR1 domain (245 to 430 aa) in AML1-ETO: deletion of this region abrogated the binding of p300 to AML1-ETO and led to loss of AML1-ETO lysine acetylation. Furthermore, loss of the region containing NHR1 domain abrogated the self-renewal properties of AML1-ETO and the activation of AML1-ETO target genes in human CD34+ cord blood cells, without affecting its differentiation-blocking activity or its ability to repress gene expression. Given the importance of the acetylation of AML1-ETO in its biological effects, we inhibited p300 function, chemically and using RNA interference; this blocked the transcriptional activation of AML1-ETO target genes, and inhibited the growth of AML1-ETO expressing AML cells in both pre-leukemic and leukemia models. All together, we have found that the acetylation of AML1-ETO via p300 is indispensable for its leukemia-promoting activity and for its ability to activate gene expression. Our work suggests that inhibition of p300 function may represent an important new anti-leukemia strategy that targets self-renewing, leukemia-initiating cells. 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: 2010
    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. 104, No. 11 ( 2004-11-16), p. 4576-4576
    Abstract: ABVD is widely considered as the gold standard treatment for advanced Hodgkin’s lymphoma (HL), although about 40% of patients relapse or do not respond to initial treatment. Recently, the HD9 trial of the German Hodgkin’s Lymphoma Study Group has shown that escalated (esc) BEACOPP regimen can achieve better disease control than ABVD, but has a high incidence of acute and long-term toxicities including high occurrence of AML/MDS. In an attempt to decrease toxicity while preserving the potential benefit of upfront intensive therapy, we conducted a pilot study, which tested the feasibility, toxicity and efficacy of combined escBEACOPP-ABVD regimen as therapy for newly diagnosed patients with high risk (IPS≥3) stage III–IV HL. Patients initially received 2 cycles of escBEACOPP followed by reevaluation with CT and gallium or PET/CT-FDG scans. When complete response (CR) or partial response (PR) was achieved, patients continued to receive 4 cycles of ABVD, while those failing to achieve a response were withdrawn from the study. Since August 2001, 21 patients entered the study and at the time of present analysis four of them are still receiving therapy. Three (18%) of 17 patients, who completed chemotherapy, received consolidative radiotherapy. Median age at diagnosis was 26 years (range 18–56) and 11 (57%) were males. Stage IV and B symptoms were evidenced in 19 (90%) and 17 (81%), respectively, and 7 (33%) had bulky mediastinal mass. Histology included nodular sclerosis in 18 patients (86%), mixed cellularity in 2 (10%) and lymphocyte predominance in 1 (5%). Following the first 2 cycles of escBEACOPP the overall response rate (CR+PR) was 100%. At the end of all therapy 15 patients (88%) were in CR, one patient in PR and only a single patient had progressive disease. With a median follow-up of 20 months no patient relapsed or died. Toxicity included WHO grade III–IV granulocytopenia in 15 patients (88%) and grade III–IV infection in one patient. Hospitalization for intravenous antibiotics was necessary in 10 patients (59%). Almost all of these events occurred during the first two cycles of escBEACOPP, while acute toxicity during the ABVD phase was mild. In conclusion, the combined escBEACOPP-ABVD regimen is well tolerated and is associated with a high CR rate when used in advanced HL patients with high IPS scores. Further follow-up is obviously required in order to determine long-term survival and late complications of this regimen.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2004
    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...