Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 32, No. 15_suppl ( 2014-05-20), p. 3505-3505
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2014
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 33, No. 3_suppl ( 2015-01-20), p. 575-575
    Abstract: 575 Background: Molecular diagnostics can identify subgroups of colorectal and other cancers that are relevant for the mode of action of new anti-cancer agents. The efficient, GCP-conform and quality assured molecular screening to identify potential study patients is one of the major challenges for targeted drug development. The EORTC has implemented a new collaborative molecular screening platform in order to facilitate these next generation trials. Methods: SPECTAcolor (Screening Patients for Efficient Clinical Trial Access in advanced colorectal cancer) is a pan European network of institutions collaborating in centralized, high-throughput screening of tumor material from patients with colorectal cancer. After informed written consent, patients are screened for molecular alterations. Molecularly annotated patients can be offered so called “downstream” clinical trials. Results: Launched in September 2013, SPECTAcolor is now initiated in 19 clinical centers in 9 countries. As of 12 September 2014, 406 patients were enrolled exceeding the expected accrual target of 300 patients in the first year. Of these 406 patients, 293 patients had their tumor block shipped to the central biobank at the Dresden University Hospital for central quality and pathological review, and core analyses. Tumor samples are being additionally analyzed using next generation sequencing for 360 key cancer alterations in cooperation with the Sanger Institute (Cambridge, UK). In the first year, the blocks were analyzed for 5 baseline biomarkers. KRAS was wild type for exon 2, 3 and 4 in 151/284 pts (53%) and mutated 133 pts (47%; 114 pts in exon 2 (40%), 8 pts in exon 3, and 11 pts in exon 4). NRAS was tested in KRAS wild type pts only; mutations were found in 14 pts (4.9%; 6 pts in exon 2 and 8 pts in exon 3). BRAF mutations, all in exon 15, were found in 18 pts (7%). PI3K mutations occurred in 41 pts (15%; 13 in exon 20 and 28 in exon 9). IHC staining was showing deficient mismatch repair in 16 patients. Conclusions: SPECTAcolor is the first pan-European and EORTC screening platform. Its successful implementation proves that a logistically complex infrastructure to run next generation trials in a multinational setting is feasible.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2015
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 3411-3411
    Abstract: Abstract 3411 Background: Dose of therapy and time to response may be different in the elderly as compared to younger patients with CML. This has been reported previously for interferon α (Berger et al., Leukemia 2003). For imatinib, contradictory results have been presented (Rosti et al. Haematologica 2007, Guliotta et al. Blood 2009). Aims: An analysis comparing dose-response relationship in patients more or less than 65 years (y) of age is warranted. Methods: We analysed the German CML-Study IV, a randomized 5-arm trial to optimize imatinib therapy by combination, dose escalation and transplantation. Patients older and younger than 65y randomized to imatinib 400 mg (IM400) or 800 mg (IM800) were compared with regard to time to hematologic, cytogenetic and molecular remissions, imatinib dose, adverse events (AEs) and overall survival (OS). Results: From July 2002 to April 2009, 1311 patients with Ph+ CML in chronic phase were randomized, 623 patients were evaluable, 311 patients for treatment with IM400 and 312 for IM800. 84 (27%) and 66 (21%), respectively, were older than 65 years. All patients were evaluable for hematologic, 578 (140 〉 65y and 438 〈 65y) for cytogenetic, and 600 (143 and 457, respectively) for molecular responses. Median age was 70y vs. 49y for IM400 and 69y vs. 46y for IM800. The median dose per day was lower for elderly patients with IM800 (517mg vs. 666mg) and the same with IM400 (400mg each). Patients' characteristics at baseline were evenly distributed in all groups regarding gender, follow-up, hemoglobin, platelet count and spleen size. Leukocyte counts were significantly lower in elderly patients (IM400: 50/nl vs. 78/nl, IM800: 36/nl vs. 94/nl). EURO score was different due to age in elderly patients (low risk: IM400: 11% vs. 43%, IM800: 14% vs. 42%; intermediate risk: IM400: 79% vs. 44% and IM800: 73% and 43%). There was no difference in cytogenetic and molecular analyses between treatment groups. With regard to efficacy, there was no difference for older patients in achieving a complete cytogenetic remission (CCR) and major molecular remission (MMR) if IM400 and IM800 were compared together. If treatment groups were analyzed separately, older patients treated with IM400 reached CCR and MMR statistically significant slower than younger patients (CCR: median 14.2 months vs. 12.1 months, p=0.019; MMR: median 18.7 months vs. 17.5 months, p=0.006). There was no difference with IM800 (CCR: median 7.7 months vs. 8.9 months, MMR: median 9.9 months vs. 10.0 months). 3y-OS for older patients 〉 65y was 94.7% and for patients 〈 65y was 96.1%. Some differences were observed in the safety analyses. 530 patients (IM400: 278, IM800: 252) were evaluated on common toxicity criteria (WHO). Some hematologic AEs were documented slightly more often in the elderly than in the younger patients: for IM400 anemia grade 1–2 (60 vs. 42%) and leukopenia grade 3–4 (5.6 vs. 1.4%) and for IM800 anemia grade 1–4 (64 vs.47% and 7.2 vs. 5.7%) and thrombocytopenia grade 3–4 (9.3 vs. 7.1%). Non hematologic AEs were more prominent in IM800 and were mainly gastrointestinal symptoms (IM400: 33 vs. 31%, IM800: 48 and 44%) and edema (IM400: 28 vs. 29%, IM800: 35 vs. 50%). There was no difference for grade 3/4 non-hematological AEs in older patients in both groups. Conclusions: Imatinib 400 mg and 800 mg are well tolerated also in the elderly. The IM800 dosage was more tolerability-adapted for the elderly, but there was no difference in reaching a CCR and MMR in contrast to the IM400 where a significantly slower response was detected in the elderly. Whether this difference is clinically relevant has yet to be determined. Updated results will be presented. Disclosures: Haferlach: MLL Munich Leukemia Laboratory: Employment, Equity Ownership, Research Funding. German CML-Study Group:Deutsche Krebshilfe: Research Funding; Novartis: Research Funding; Roche: Research Funding; BMBF: Research Funding; Essex: Research Funding.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 357-357
    Abstract: Abstract 357 Treatment of CML with imatinib of 400 mg can be unsatisfactory. Treatment optimization is warranted. The German CML-Study group has therefore conducted a randomized study comparing imatinib 800 mg vs 400 mg vs 400 mg + IFN. A significantly faster achievement of MMR at 12 months has been observed with imatinib 800 mg in a tolerability adapted manner and MMR by 12 months has been found to translate into better overall survival. Since stable CMR has been associated with durable off-treatment remissions we sought to analyse the impact of tolerability-adapted imatinib 800 mg on CMR and survival. Standardized determinations of molecular response and evaluation of its impact on outcome are goals of CML-Study IV. CMR4 is defined as a BCR-ABL/ABL ratio of 〈 0,01 on the International Scale. From July 2002 – April 30, 2009 1022 newly diagnosed patients with CML in chronic phase were randomized, 1012 were evaluable (338 with imatinib 800 mg, 324 with imatinib 400 mg, 350 with imatinib plus IFN). Median observation time was 40 months. The median average daily imatinib doses were 628 mg in the 800 mg arm and 400 mg in the 400 mg based arms. The actual median daily doses in the 800 mg arm per 3-months periods were: 555 mg, 737 mg, 613 mg, 600 mg, and 600 mg thereafter, reflecting the run–in period with imatinib 400 mg for 6 weeks in the first period and the adaptation to tolerability from the third 3-months period onwards. Median daily imatinib doses in the 400 mg arms were 400 mg throughout. Adaptation of imatinib dose in the 800 mg arm according to tolerability is reflected by similar higher-grade adverse events rates (WHO grades 3 and 4) with all treatments. Significantly higher remission rates were achieved with imatinib 800 mg by 12 months. The cumulative incidences of CCR by 12 months were 63% [95%CI:56.4-67.9] with imatinib 800 mg vs 50% [95%CI:43.0-54.5] with the two 400 mg arms. The cumulative incidences of MMR by 12 months were 54.8% [95%CI:48.7-59.7] with imatinib 800 mg vs 30.8% [95%CI:26.6-36.1] with imatinib 400 mg vs 34.7% [95%CI:29.0-39.2] with imatinib + IFN. The cumulative incidences of CMR4 compared with the MMR incidences over the first 36 months are shown in Table 1. Imatinib 800 mg shows superior CMR4 rates over the entire 36 months period, CMR4 is reached significantly faster with imatinib 800 mg as compared to the 400 mg arms. The CMR4 rates reach 56.8% by 36 months [95%CI:49.4-63.5] as compared to 45.5% with imatinib 400 mg [95%CI:38.7-51.0] and 40.5% with imatinib plus IFN [95%CI:34.6-46.3] . Most patients have stable CMR4 over the entire period. Time after start of treat-ment (months) Cumulative incidences MMR(%) CMR4 (%) IM400 n=306 D IM800 n=328 D IM400 +IFN n=336 IM400 n=306 D IM800 n=328 D IM400 +IFN n=336 6 8.6 9.5 18.1 9.7 8.4 3 0.7 3.7 1.3 2.4 12 30.8 24.0 54.8 20.1 34.7 7.5 12.3 19.8 7.4 12.4 18 50.3 18.1 68.4 14.3 54.1 21.2 12.2 33.4 9.8 23.6 24 63 13.0 76.0 13.2 62.8 30.7 12.3 43 13 30.0 36 79.3 2.3 81.6 10.9 70.7 45.5 11.3 56.8 16.3 40.5 In summary, superior CMR4 rates are achieved with high-dose imatinib adapted to good tolerability, and more patients in the tolerability-adapted 800 mg arm have stable CMR4 qualifying for treatment discontinuation as compared to the 400 mg based arms. With improved application imatinib remains first choice for early CML. Disclosures: Koschmieder: Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; BMS: Membership on an entity's Board of Directors or advisory committees. Schnittger:MLL Munich Leukemia Laboratory: Employment, Equity Ownership. German CML-Study Group:Deutsche Krebshilfe: Research Funding; Novartis: Research Funding; Roche: Research Funding; BMBF: Research Funding; Essex: Research Funding.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 785-785
    Abstract: Abstract 785 Background: FLT3 internal tandem duplications (FLT3-ITD) occur in about 25% of acute myeloid leukemia (AML), are associated with cooperating gene mutations (NPM1, DNMT3A), and confer an adverse prognosis. Several studies have indicated that the unfavorable impact of FLT3-ITD is influenced by a number of factors, such as the mutant to wild-type ratio (allelic ratio), insertion site of FLT3-ITD in the beta1 sheet of the tyrosine kinase domain 1, and the molecular background of cooperating mutations. Aims: To evaluate the relative impact of FLT3-ITD allelic ratio and insertion site, as well as cooperating genetic lesions on prognosis and treatment decision making in a large cohort of homogeneously treated younger adult patients. Methods: The basis of the study were 2377 younger adults (median age, 48 years; range, 16–62 years) with newly diagnosed AML enrolled on three prospective treatment trials of the German-Austrian AML Study Group (AMLSG) between 1993 and 2008. Patients with acute promyelocytic leukemia (n=99), core-binding factor AML (n=279) and AML with adverse-risk cytogenetics (n=436) according to the European LeukemiaNet recommendations were excluded. Based on material availability, the presence of FLT3-ITD could be analyzed in 1414 patients; NPM1 and DNMT3A mutational status was available in 97% and 84% of the patients, respectively. In FLT3-ITD positive AML (n=394), the allelic ratio, determined by Genescan-based fragment-length analysis, was available in 86% and the insertion site in 72%. Allogeneic hematopoietic stem cell transplantation (HSCT) in first complete remission was performed in 41% and 29% of FLT3-ITD positive and negative patients, respectively. Results: We first evaluated the prognostic impact of the different FLT3-ITD characteristics within the subgroup of FLT3-ITD positive patients. The allelic ratio was categorized into quartiles ranging from low to high. For the endpoints event-free (EFS), relapse-free (RFS) and overall survival (OS), only the fourth quartile with the highest allelic ratio showed a prognostic impact for all endpoints, whereas no difference was identified between the other three quartiles. For further analyses, the allelic ratio was dichotomized comparing the fourth quartile versus the other three quartiles. FLT3-ITD insertion site in the beta1 sheet was significantly associated with an unfavorable outcome for all endpoints. Additionally, FLT3-ITD size was directly correlated with the insertion site: the more C-terminal the ITD inserted in the FLT3 gene the longer the FLT3-ITD size. There was no prognostic impact of FLT3-ITD size neither as continuous nor as quartile-categorized variable. Multiple FLT3-ITDs, present in 13% of AMLs, were associated with an unfavorable prognosis. The presence of either NPM1 and/or DNMT3A mutations in FLT3-ITD positive patients did not alter the original FLT3 prognosis. In multivariable models for the endpoint OS of the total cohort of intermediate-risk AML, an independent prognostic impact beyond the variable FLT3-ITD was shown for the allelic ratio (fourth quartile) [HR, 1.4; p=0.037] and in trend for insertion site in the beta1 sheet [HR, 1.33; p=0.06] . Survival of patients exhibiting a high allelic ratio (n=43) or insertion site in the beta1 sheet (n=60) was comparable, with a median of 10 and 13 months and 4-year survival of 19% and 24%, respectively. Of note, outcome of patients with both high allelic ratio and insertion site in the beta1 sheet (n=21) was very poor with a median OS of 10 months and 4-year OS of 5%, respectively. In patients with FLT3-ITD positive AML without these unfavorable factors (n=144), median and 4-year OS were 15 months and 42%, respectively. Of note, a clear benefit of allogeneic HSCT in first CR was only seen in FLT3-ITD positive patients without these two unfavorable factors, with a 4-year OS of 63%. In comparison, the 4-year OS of the same subgroup of patients achieving a CR after induction therapy without proceeding to allogeneic HSCT during first CR was 35%. In contrast, outcome in patients with high allelic ratio and/or insertion site in the beta1 sheet remained poor despite allogeneic HSCT in first CR. Conclusion: High FLT3-ITD allelic ratio and ITD insertion site in the beta1 sheet presented as prognostic indicators for poor outcome in patients with the presence of a FLT3-ITD. Only patients without these unfavorable FLT3-ITD features significantly benefitted from allogeneic HSCT. Disclosures: Schlenk: Roche: Research Funding; Pfizer: Research Funding; Amgen: 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Society of Clinical Oncology (ASCO) ; 2020
    In:  Journal of Clinical Oncology Vol. 38, No. 6_suppl ( 2020-02-20), p. 705-705
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 38, No. 6_suppl ( 2020-02-20), p. 705-705
    Abstract: 705 Background: Using sunitinib (SUN) as the comparator, two out of three recent phase III clinical trials have shown an overall survival (OS) benefit in the alternative treatment arm (Checkmate 214 (C), Keynote 426 (K)). The Javelin 101(J) trial has yet to show a significant improvement. Methods: A cross-study comparison of OS for the sunitinib arms of C, K and J was carried out. Special focus was given to risk group stratification and subsequent therapies. Data from the SUN pivotal trial (SP) was also taken into account. Results: Across the C, K and SP trials, OS was similar at approximately 78%. Despite a lower percentage of favourable risk patients compared to K and SP, an OS of 82% was observed in J. At 18 months, OS survival curves split ranging from 65% ©, 72.1% (K) to 76% (J). OS in the SUN arm in J at 18 months was comparable to OS of Nivolumab/Ipilimumab in C (78%). The rate of subsequent therapies ranged from 34.3% (K), 39.2% (J) to 54% (C) in the SUN arms. At the time of the SP trial almost no subsequent therapy options existed. In the experimental arms, the rate of subsequent therapies was 51.8% ©, 20.5% (K) and 20.8% (J). The existing data give no clear evidence of a correlation between subsequent therapies and OS. However, an analysis of the subsequent therapies/patients discontinuing therapy ratio (ST/DIS) and type of subsequent therapy (PD1/L1 directed or therapy with proven OS benefit (pOS) in randomized trials) in the SUN arms may be more conclusive (see table). The rate of subsequent therapies (ST/DIS) as well as PD(L)1 or pOS therapies were highest in J, followed by K and C. Conclusions: Despite the difficulties of a cross trial comparison, this data should raise awareness of the influence of a more intense subsequent therapy. It points to the necessity to standardize subsequent therapies in 1st line trials if OS is a primary study aim. Given this analysis, it may be pertinent to ask if an optimal or near optimal subsequent therapy following SUN might be equal to a 1 st line IO/IO therapy with regards to OS.[Table: see text]
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2020
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    Ovid Technologies (Wolters Kluwer Health) ; 1999
    In:  Anti-Cancer Drugs Vol. 10 ( 1999-11), p. S5-S12
    In: Anti-Cancer Drugs, Ovid Technologies (Wolters Kluwer Health), Vol. 10 ( 1999-11), p. S5-S12
    Type of Medium: Online Resource
    ISSN: 0959-4973
    Language: English
    Publisher: Ovid Technologies (Wolters Kluwer Health)
    Publication Date: 1999
    detail.hit.zdb_id: 2025803-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 412-412
    Abstract: Abstract 412 Background: Somatic mutations of the ASXL1 (Additional Sex Comb-Like 1) gene on chromosome 20q11.1 were identified in various myeloid malignancies with the highest incidence reported in CMML (∼40%) and lower frequencies in MDS, AML, CML, and myeloproliferative neoplasia. The ASXL1 protein has been suggested to act as chromatin modifier and the highly conserved C-terminal plant homeo-domain (PHD) finger is presumably critical for its function. ASXL1 mutations cluster in exon 12 and are mainly frameshift mutations predicted to remove the PHD domain. We and others (Paschka et al., Haematologica 2011;96(s2);425; Chou et al., Blood 2010;116:4086–94) have recently reported first results on the unfavorable prognostic impact of ASXL1 mutations in AML. However, the clinical relevance of these mutations still needs to be elucidated in larger AML cohorts. Methods: Mutational analyses of ASXL1 were performed on diagnostic samples from 1429 patients with AML aged 18 to 61 years. All patients were intensively treated on one of two AMLSG trials [AML HD98A (n=745), Schlenk et al., J Clin Oncol. 2010;28:4642–8; AMLSG 07–04(n=684), NCT00151242]. GeneScan-based fragment analysis of several amplicons spanning ASXL1 exon 12 was used to screen for ASXL1 mutations. Samples showing altered GeneScan profiles were amplified in a second PCR reaction, and the amplicons were sequenced to confirm the mutation and to determine the mutation type. Patients were also assessed for the presence of NPM1, FLT3 (ITD and TKD), CEBPA, IDH1/2, RUNX1, and DNMT3A mutations by standard PCR-based methods. Results: ASXL1 mutations were detected in 90 (5.9%) of 1429 patients. All mutations were heterozygous frameshifts predicted to cause loss of the PHD finger. The most common mutation was a duplication of guanine at position 1934 (c.1934dup) identified in 59% (53/90) of the mutated cases. Three other ASXL1 mutations were detected in more than one patient: c.1900_1922del (n=16), c.1934del (n=5), c.1960dup (n=3). The majority of mutations (91%) clustered within or around a glycine-rich protein domain spanning amino acids 642–685. Patients with ASXL1 mutations were older (P 〈 .0001), more frequently males (P=.03), and they more frequently had secondary AML (P=.02). They also showed lower values for WBC (P=.01), bone marrow (P=.0002) and circulating (P=.001) blasts, and LDH serum levels (P=.007). ASXL1 mutations were more frequent in patients exhibiting intermediate-risk cytogenetics (P=.06). Among the ASXL1 mutated cases, 42% were cytogenetically normal (CN), 23% had other intermediate-risk cytogenetics, 26% had high-risk, and 9% low-risk [t(8;21) n=7, t(15;17) n=1, no inv(16)/t(16;16)] cytogenetics. ASXL1 mutations were associated with RUNX1 (P 〈 .001) and IDH2R140 mutations (P=.006). In contrast, ASXL1 mutations were less frequent in patients with NPM1 (P 〈 .001), FLT3-ITD (P 〈 .001), and DNMT3A (P=.02) mutations. The median follow-up for survival was 4.8 years. Patients with an ASXL1 mutation had an inferior complete remission (CR) rate compared with ASXL1 wildtype patients (57% vs 74%; P 〈 .001); the same was true for the subset of CN-AML (58% vs 77%; P=.04). In both, the entire cohort (P=.04) and in CN-AML (P=.07) ASXL1 mutations were associated with a worse event-free-survival, whereas no impact of these mutations was observed on relapse-free-survival. Patients with ASXL1 mutation had a shorter overall survival (OS) compared to those with ASXL1 wildtype (P=.002; 4-year OS rates, 32% vs 47%). The adverse impact of ASXL1 mutations on OS was also present in the subgroup of CN-AML (P=.009; 4-year OS rates, 33% vs 48%). In multivariable analysis, ASXL1 mutation was in trend an unfavorable factor for OS in CN-AML (hazard ratio: 1.46; P=.087). Conclusions: ASXL1 mutations were identified in ∼6% of younger AML patients and were associated with intermediate-risk cytogenetics. ASXL1 mutations frequently occurred with RUNX1 and IDH2R140 mutations, and were less frequent in NPM1 and FLT3-ITD mutated AML. ASXL1 mutations were associated with lower CR rate and inferior OS. The exact role of ASXL1 in normal hematopoiesis still needs to be defined in functional studies as well as the potential role of mutated ASXL1 protein in mediating resistance to chemotherapy. Disclosures: Kündgen: Celgene: Honoraria; Novartis: Honoraria.
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 14, No. 1 ( 2008-01-01), p. 215-223
    Abstract: Purpose: To determine the recommended dose (RD) of EKB-569, an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, in combination with FOLFIRI chemotherapy in patients with metastatic colorectal cancer (mCRC). Methods: Patients with previously untreated mCRC received FOLFIRI and EKB-569 at doses of 10, 25, 50, and 75 mg/day (EKB started on day 3). Three sequential skin biopsies were obtained in selected patients to assess the pharmacodynamic effects on EGFR signaling of FOLFIRI alone and with EKB-569. Complete pharmacokinetic sampling and tumor biopsies, when feasible, were conducted. Results: Forty-seven patients were enrolled. Dose-limiting toxicities (grade 3 diarrhea or asthenia) were observed in 1/7 patients at 50 mg EKB-569 and in 2/3 at 75 mg. Two additional dose levels (35 mg EKB-569/day and 50 mg EKB-569/day plus modified FOLFIRI) were evaluated. The RD was 25 mg EKB-569/full dose FOLFIRI. Grades 3 to 4 toxicities in & gt;10% of patients were diarrhea (30%), neutropenia (21%), and asthenia (17%). Twenty-one patients had an objective response [48%; 95% confidence interval (95% CI), 32-65%]. The median time to tumor progression was 7.7 months. At the RD, EKB-569 resulted in complete inhibition of phosphorylated EGFR (pEGFR) and downstream receptor signaling in skin and tumor samples. FOLFIRI alone did not affect pEGFR, but inhibited epidermal proliferation and activated mitogen-activated protein kinase (MAPK) and induced p27 expression in the skin. Conclusion: The RD of EKB-569 is 25 mg/day when combined with FOLFIRI and results in complete EGFR inhibition. Chemotherapy alone interferes with pharmacodynamic markers, an observation to be taken into account in future studies of targeted agents with chemotherapy.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2008
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 80-80
    Abstract: Abstract 80 Background: Mutations in the nucleophosmin-1 gene (NPM1) are the most common genetic abnormalities in acute myeloid leukemia (AML) and define a provisional AML entity in the current WHO classification. In a retrospective biomarker study within a randomized trial of older patients with AML, we demonstrated that patients with mutated NPM1 and absence of a FLT3 internal tandem duplication (ITD) benefit from all-trans retinoic acid (ATRA) as adjunct to conventional chemotherapy (Schlenk et al. Haematologica 2009;94:54–69). Aims: To evaluate the impact of ATRA in combination with conventional chemotherapy on outcome, and to assess the NPM1 mutational status as predictive marker for response to this therapy in younger adult patients with AML entered in the prospective randomized controlled treatment trial AMLSG 07-04 (ClinicalTrials.gov Identifier: NCT00151242). Methods: Patients (18 to 60 years of age) were accrued between August 2004 and August 2009. They were randomized up-front for open-label treatment with ATRA. Induction therapy consisted of two cycles of ICE (idarubicin 12mg/m2, day 1,3,5 [in induction II reduced to d 1, 3]; cytarabine 100mg/m2 continuous i.v., day 1 to 7; etoposide 100mg/m2, day 1–3). For consolidation therapy, patients with high-risk AML, defined either by high-risk cytogenetics or induction failure, were assigned to receive allogeneic hematopoietic stem cell transplantation (HSCT) from a matched related (MRD) or unrelated donor (MUD). Starting from November 2006, AML with FLT3-ITD was also categorized as high-risk. All other patients were assigned to 3 cycles of high-dose cytarabine (HiDAC; 18g/m2 per cycle). In all but patients with core-binding-factor AML an allogeneic HSCT was intended when a MRD was available. During induction cycles, ATRA was given in a dosage of 45mg/m2 from day 6 to 8, and 15mg/m2 from day 9 to 21; and during HiDAC cycles in a dosage of 15mg/m2 from day 6 to 28. The primary end points of the study were event-free survival (EFS) and rate of complete remission (CR) after induction therapy; secondary end points were, relapse-free (RFS) and overall survival (OS). For survival analyses, patients receiving an allogeneic HSCT in first CR were censored at the date of transplantation. Forty patients were treated either with or without ATRA despite being randomized in the opposite treatment arm; predictive marker analyses were performed on a per protocol basis excluding those patients. Results: A total of 1112 patients were randomized, 562 (per protocol 542) in the standard arm, and 550 (per protocol 530) in the investigational arm with ATRA. Median follow-up was 3.3 years. NPM1 mutational status was assessed in 1018 patients (92%) and a mutation was identified in 289 (28%) patients. Pretreatment patient characteristics at diagnosis were well balanced between the standard and the ATRA-arm of the study, except for higher white blood counts (WBC) in the standard arm (median, 16.1/nl vs. 8.9/nl, p=0.001). The CR-rate was significantly increased in NPM1-mutated AML by ATRA (OR, 2.20; p=0.05), independent of the FLT3-ITD status (OR, 0.66; p=0.33); there was no effect of ATRA in NPM1-wild-type AML (OR, 1.00; p=0.99). Multivariable analyses on EFS revealed a significant risk reduction in NPM1-mutated AML by ATRA (hazard ratio [HR], 0.65; p=0.02), whereas there was no effect of ATRA in NPM1-wild-type AML (HR, 0.99; p=0.95). Other significant factors in NPM1-mutated AML were IDH1R132 mutation (HR, 1.72; p=0.04), IDH2R140 mutation (HR,1.73; p=0.03), FLT3-ITD (HR, 1.55; p=0.04), log-transformed WBC (HR, 1.47; p=0.03), and in NPM1-wild-type AML IDH2R172 mutation (HR,1.82; p=0.03), FLT3-ITD (HR, 1.56; p=0.002), logarithm of WBC (HR, 1.20; p=0.02), male gender (HR, 1.34; p=0.003), cytogenetic risk (p 〈 0.001; HR (high-risk vs intermediate risk), 2.18; HR (low-risk vs intermediate risk) 0.31). ATRA had no influence on the cumulative incidence of relapse. OS of patients treated with ATRA (n=549) was significantly better (p=0.02) compared with that of patients not treated with ATRA (n=562). Rates and severity of toxicities were similar in both treatment arms. Conclusions: ATRA in addition to conventional chemotherapy significantly improved response to induction therapy and EFS in NPM1 mutated-AML, as well as OS in the whole cohort of younger adult patients with AML. NPM1 mutation was confirmed as a predictive factor for response to this combination therapy. Disclosures: Schlenk: Roche GmbH: Research Funding; Amgen GmbH: Research Funding; Pfizer GmbH: Research Funding. Krauter:Novartis: Consultancy, Honoraria. Kuendgen:Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees. Fiedler:Pfizer: 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages