Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 114, No. 22 ( 2009-11-20), p. 1055-1055
    Abstract: Abstract 1055 Poster Board I-77 The combination of the histone deacetylase inhibitor vorinostat with Ida and ara-C has synergistic antileukemia activity in a sequence dependent fashion (Blood 2006;108:1174-82; CCR 2009;15:1698-707). The combination of high-dose vorinostat and Ida is safe and active in patients (pts) with relapsed/refractory AML (Kadia et al, submitted). Based on this, we designed a phase II study of vorinostat followed by Ida and ara-C in pts with AML or MDS. Eligibility criteria included previously untreated AML (excluding pts with APL), or int-2 or high risk MDS, age 〉 65 years, and adequate renal, hepatic and cardiac functions as well as performance status. Schedule for induction therapy is vorinostat 500 mg orally (po) three times a day (TID) for 3 days (days 1 to 3), Ida 12 mg/m2 IV daily x 3 (days 4 to 6) and ara-C (1.5 gm/m2 as a continuous (CI) 24 hours infusion on days 4 to 7). Five cycles of consolidation therapy are planned with same dases of vorinostat (days 1 to 3) but decreased doses of Ida (8 mg/m2 days 4 and 5) and ara-C (0.75 gm/m2 CI x 24 hours on days 4 to 6). Maintenance therapy consists of vorinostat 200 mg po TID daily x 14 days every 28 days for 12 cycles. The study is designed to stop early based on a composite endpoint and predefined stopping rules of progression free survival (PFS) (the study is to stop if a PFS of at least 7 months is not expected to be achieved), toxicity and response rates compared to standard Ida and Ara-C (IA) combination at MD Anderson. Planned interim analyses are performed every 6 months by an independent statistician to monitor the above endpoints. To test the safety of the combination, this study included a “reversed” phase 1 run-in phase starting at the maximal dose of vorinostat planned (500 mg po qd tid x 3 days). Three patients were treated at this dose with no excess toxicity and 3 of 3 pts achieved a response. After this, the phase II portion of the study started. A maximum of 75 pts are to be treated if no stopping rule is met. Currently 52 pts have been enrolled and 45, all with AML, are evaluable for response (median age 53 yeas (range 19-65), 21 (46%) female, 9 (20%) with Flt-3 mutations, 30 (66%) with abnormal cytogenetics, 24 (53%) with t-AML or antecendent MDS, median WBC was 5.5 (range 0.9-110.6). In total 43 (95%) pts had poor risk AML as defined by poor CG or Flt-3 or t-AML/MDS. Induction mortality was 2 (4%), Complete remission (CR) after was one course of therapy was achieved in 35 pts and 1 pt achieved a CRP (incomplete platelet recovery) for an overall response rate of 80%. Seven (15%) pts did not respond to therapy. CR by molecular characteristics are: diploid 13 of 15 (86%), Flt-3 9 of 9 (100%), abnormal CG (20 of 30, 66%). No excess toxicity with the addition of vorinostat has been observed compared to standard IA. Common complications were related to myelosuppresion typical of IA. No excess nausea or vomiting or diarrhea was observed. With a median follow up of 4.2 months (range 0.7 – 13 months), median OS has not been reached and median DFS was 9.5 months. The study continues to be open after two interim analysis, as the PFS is higher than 7 months (expected from standard IA). The median number of courses of consolidation therapy is 2+ (range 0-5) and 4 pts are receiving maintenance therapy without excess toxicity. Markers of induction of autophagy (LC3-II), ROS activiation and NF-Kb signaling are upregulated in pts receiving this therapy. This study indicates that the combination of vorinostat, Ida and ara-C is safe and active in AML. The role of vorinostat in Flt-3 postive leukemias needs to be further studied. A randomized study of Ida and ara-C with or without vorinostat needs to be performed to establish the role of vorinostat in front-line AML therapy. At the current accrual rate, this study will be completed by the ASH meeting. Disclosures: Garcia-Manero: Merck: Research Funding. Off Label Use: Vorinostat is not approved for use in AML or MDS.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    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 ...
  • 2
    In: Blood, American Society of Hematology, Vol. 118, No. 21 ( 2011-11-18), p. 763-763
    Abstract: Abstract 763 The combination of vorinostat, a panhistone deacetylase inhibitor with activity in relapsed AML (Blood 2008;111:1060), with idarubicin and ara-C has synergistic antileukemia activity in a sequence dependent fashion were optimal effect is observed when vorinostat precedes ara-C (Blood 2006;108:1174 and CCR 2009;15:1698). In a phase I trial, we documented the maximal tolerated dose of vorinostat to be 500 mg orally three times a day for 3 days with standard dose idarubicin (BJH 2010; 150:72). Based on this we performed a phase II trial of vorinostat with idarubicin and ara-C in front line AML and higher risk MDS. Patients ages 15 to 65 years with appropriate organ function and no core binding factor abnormality were candidates. Induction therapy was vorinostat 500 mg orally three times a day (days 1 to 3), idarubin 12 mg/m2 IV daily × 3 (days 4 to 6), cytarabine 1.5 gm/m2 IV as a continuous infusion daily for 3 or 4 days (days 4 to 7). Patients in remission could be treated with 5 cycles of consolidation therapy with lower doses of the combination and up to 12 months of maintenance with single agent vorinostat. The study was designed to stop early if excess toxicity or low probability of event free survival (EFS) 〉 28 weeks were likely. After a 3 patient run-in phase, 75 patients were treated. Median age was 52 (19–65) years, 29 (39%) were diploid and 11 (15%) were Flt-3 ITD. Therapy was well tolerated: no excess vorinostat related toxicity was observed. Induction mortality was 4%. Common toxicities included: diarrhea (72%), nausea and vomiting (65%), and skin toxicity (38%). No cardiac toxicity was observed. CR was documented in 57 (76%) patients and CRp in additional 7 (9%) for an overall response rate (ORR) of 85% (95% CI 0.75–0.91). No differences in response were observed in patients younger or older than 60 years. Seven (9%) patients received 2 cycles of induction therapy and 4 (44%) responded including 2 CR and 2 CRps. When analyzed based on molecular or cytogenetic features, 10 of the 11 patients with Flt-3 ITD achieved a CR and 1 a CRp (ORR 100%). All 6 NPM1 mut/Flt-3 wild type (WT) patients achieved CR. Presence of Ras mutations had no impact on response. Of the diploid patients, 25 (86%) achieved a CR and 2 a CRp (7%) (ORR 93%). Patients with other cytogenetic alteration excluding −5/–7 had an ORR of 93% including 23 (79%) CR and 4 CRp (14%). Only 9 (53%) patients with −5/−7 achieved a CR and 1 (6%) a CRp for an ORR of 64% (p=0.018). Responses were significantly higher than with standard IA therapy. With a median follow up of 82 weeks (5 to 132 weeks), the median OS for the whole group was 82 (range 3–134) and EFS 47 (3–134) weeks. Median OS and EFS for patients with Flt-3 ITD were 91 (range 6 to 134) and 66 (6–134) weeks. There was a trend towards better survival in the Flt-3 ITD patients versus the unmutated group (p=0.067). Patients with diploid cytogenetics had a median OS and EFS of 105 (5–134) and 68 weeks (5–134). There was a trend towards better survival in the diploid patients versus non-diploid patients (p=0.09). Survival was poorer in patients with −5/−7: OS was 34 weeks (range 3–92) and EFS 14 (3–92). Non-diploid non −5/−7 patients had a median OS of 92 weeks (5–119) and EFS was not reached (NR) (5–119). Nineteen patients (25%) were transplanted in CR#1. All patients had achieved CR/CRp before SCT. Eleven (57%) received an unrelated donor including 2 umbilical cords and two 1 antigen mismatch. Fifteen patients (79%) received ablative preparative regimens. Median OS and EFS had not been reached in the 19 patients transplanted. Robust induction of histone H3 acetylation was documented in 2 patients (2%). No induction of beclin was observed in any of the patients. Baseline levels of NRF2 were elevated in 6 of 34 patients (17%), CYBB 21 of 34 (61%), FOXO3 5 of 34 (14%), SOD1 11 of 34 (32%), SOD2 5 of 34 (14%), GST-PI 2 of 34% (6%), IL6 3 of 49 (6%), TIMP1 7 of 49 (14%), MMP9 4 of 49 (8%) and p38MAPK in 3 of 49 (6%). mRNA upregulation occurred in all genes starting as early as day 3 of therapy. Baseline levels of NRF2 and CYBB were associated with longer survival. No association between age or cytogenetics were found with NRF2 or CYBB. In summary, the combination of vorinostat with IA is associated with high induction response rates. Toxicity and EFS rules were not met and the study beat expectations. Further studies comparing it to standard IA or “7+3” programs and in patients with Flt-3 ITD should be proposed. 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: 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 ...
  • 3
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 444-444
    Abstract: Epigenetic alterations are common in leukemia. MGCD0103 in an oral isotype-selective HDAC inhibitor that synergizes in vitro with the DNA methyltransferase inhibitor 5-azacitidine (Vidaza, Pharmion). Both agents have single-agent clinical activity in MDS and AML (Garcia-Manero, ASCO, 2006 & Silverman, JCO, 2002). We have developed a Phase I/II study of 5-azacitidine in combination with MGCD0103 in patients with AML and MDS. Patients with MDS (≥10% marrow blasts), relapsed/refractory AML, or untreated elderly patients with AML were eligible. Adequate performance status, renal and hepatic functions were required. 5-azacitidine was administered at its approved dose/schedule: 75 mg/m2 SC daily for the first 7 days of a 28 day cycle. MGCD0103 was administered as a flat dose orally three-times a week starting on the 5th day of 5-azacitidine administration. The phase I portion of the study design followed a classic “3+3” model and only MGCD0103 was dose escalated. The phase II portion targeted a 30% response rate. Final data from the Phase I and II portions of the study will be presented at the Meeting. Five dose levels of MGCD0103 have been evaluated: 35, 60, 90, 110 and 135 mg. At current data cut-off, 37 patients registered in the study were fully evaluable: median age was 67 (range 27–85); 31 patients had AML and 6 MDS. A total of 97 cycles were administered to date, mean = 2.6 (range 1–12). Dose limiting toxicities included nausea, vomiting, anorexia, diarrhea and dehydration which appear similar to dose limiting toxicities for MGCD0103 alone. The MTD of MGCD0103 was initially determined to be 110 mg, however, upon cohort expansion, this dose level was associated with excess toxicity and the starting dose was decreased to 90 mg. Eleven (30%) patients have achieved response: 4 CR, 5 CR-i, and 2 PR. Of these 11 patients, 6 continue on study with mean duration on study of 7 cycles. Of the 5 patients discontinued, 3 discontinued due to SAEs, 1 due to progressive disease and 1 to undergo transplantation. Of the 27 patients at the phase II dose levels of 90 and 110mg, 10 achieved a response (37%; same rate at both doses). Preliminary response data are available at the time of abstract preparation for 13 additional patients, revealing 4 with CR (one of which had 1% residual peripheral blast) and 3 with CR-I for a response rate of 53% in this subset. MGCD0103 pharmacokinetics were not affected by 5-azacitidine. Likewise, co-administration of MGCD0103 had no impact on the pharmacokinetics of 5-azacitidine. A majority of patients exhibited a substantial reduction in PBMC HDAC activity during treatment with the combination. Analysis of DNA methylation is ongoing. In conclusion, the combination of 5-azacitidine with MGCD0103 is safe in patients with advanced AML/MDS and has clinical activity potentially superior to that expected with 5-azacitidine alone in this patient population. These results form the bases of a planned randomized study of 5-azacitidine with or without MGCD0103 in AML and MDS.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    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. 112, No. 4 ( 2008-08-15), p. 981-989
    Abstract: MGCD0103 is an isotype-selective inhibitor of histone deacetylases (HDACs) targeted to isoforms 1, 2, 3, and 11. In a phase 1 study in patients with leukemia or myelodysplastic syndromes (MDS), MGCD0103 was administered orally 3 times weekly without interruption. Twenty-nine patients with a median age of 62 years (range, 32-84 years) were enrolled at planned dose levels (20, 40, and 80 mg/m2). The majority of patients (76%) had acute myelogenous leukemia (AML). In all, 24 (83%) of 29 patients had received 1 or more prior chemotherapies (range, 0-5), and 18 (62%) of 29 patients had abnormal cytogenetics. The maximum tolerated dose was determined to be 60 mg/m2, with dose-limiting toxicities (DLTs) of fatigue, nausea, vomiting, and diarrhea observed at higher doses. Three patients achieved a complete bone marrow response (blasts ≤ 5%). Pharmacokinetic analyses indicated absorption of MGCD0103 within 1 hour and an elimination half-life in plasma of 9 (± 2) hours. Exposure to MGCD0103 was proportional to dose up to 60 mg/m2. Analysis of peripheral white cells demonstrated induction of histone acetylation and dose-dependent inhibition of HDAC enzyme activity. In summary, MGCD0103 was safe and had antileukemia activity that was mechanism based in patients with advanced leukemia.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    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. 114, No. 22 ( 2009-11-20), p. 119-119
    Abstract: Abstract 119 The role of hypomethylating agents is well established in patients with higher risk MDS. The activity and safety of these agents in patients with lower risk (IPSS low or int-1) is not well understood. We hypothesized based on prior in vitro data and cumulative experience with different routes of administration that subcutaneous (SQ) administration of decitabine (5-aza-2'-deoxycitidine) administered either daily for 3 days or weekly times 3 both every 4 weeks will be safe and active in patients with lower risk MDS. To test this hypothesis, we designed a phase II “play the winner” randomized study to compare the daily versus the weekly schedule of decitabine at a daily dose of 20 mg/m2 SQ. Operating characteristics of the design include a 1:1 randomization procedure with the first 40 patients (pts) and after that an adaptive procedure were pts are allocated based on preceding response characteristics. A maximum of 80 pts are plan to be treated on the study. Eligibility criteria include age older than 18 years with low or int-1 MDS by IPSS, adequate performance status and renal and hepatic functions. Pts that have received prior decitabine or 5-azacitidine are excluded. Pts that have received growth factors, lenalidomide or other investigational agents are required a wash out period of 30 days. Baseline transfusion requirements are recorded as baseline and during therapy. At the present time based on the data cut-off by July 10, 2009, 43 pts have been enrolled and treated on study with 22 on the daily times 3 arm and 21 on the weekly x 3 arm. Pt characteristics are: median age 70 years (range 32-87), median time from diagnosis 2.2 months (range 0-64), 36 (84%) pts with de novo disease, 10 (23%) with IPSS low and 32 (74%) with int-1, median WBC 5.6(range 0.6-26), Hgb 9.6 (range 4.9-14.4, platelets 97 (6-442), diploid cytogenetics 30 (70%), 13 pts (30%) with others (-Y, del5, del20, +8, others). Pt characteristics are well balanced between both arms. Of the 43 pts evaluable for response and using IWG 06 response criteria, 4 (9%) pts achieved complete remissions (CR): 3 (14%) in the daily schedule and 1 (5%) in the weekly. Two marrow responses (5%), 1 partial response (2%) and 4 hematological improvements (10%) that were equally distributed between both arms were also documented. The overall response rate is 25%, 32% for the daily arm and 19% for the weekly (p=0.3). The media number of course administered so far is 4+ (range 1-11+). The median time to initial and best response is 2 cycles (ranges 2 and 2-4 respectively). No significant non-hematological toxicity has been observed with minimal grade 3 or 4 toxicity with either arm. Mortality during the first 8 weeks due to treatment complications has been 0%. Three pts have transformed to AML including 2 pts that have died due to disease progression. All in the weekly arm. Based on data cut-off by July 10, 2009, 35 pts (81%) remain on study. With a median follow up of 4.5 months (range 0.9-10), median overall survival has not been reached. Despite the trend to increase response rates and transformation with the weekly arm, at the present time no significant advantage to any of the arms has been documented . As a pharmacodynamic end point, induction of global DNA hypomethlytion using the LINE bisulfite pyrosequencing assay has been measured in 8 consenting patients. Induction of global hypomethylation is observed (50% of pts) pts but the data is limited to establish relationships between arm and/or response. In summary, these initial results indicate that lower dose subcutaneous schedules of decitabine in patients with low or int-1 disease are safe and active with significant less myelotoxicity, and related complications, compared to standard 5-day IV schedules of decitabine used in higher risk disease. Disclosures: Garcia-Manero: Celgene: Research Funding; Eisai: Research Funding. Chen:Eisai: Employment. Stein:Eisai: Employment.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2009
    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
    In: Blood, American Society of Hematology, Vol. 110, No. 11 ( 2007-11-16), p. 1842-1842
    Abstract: Histone deacetylase inhibitors (HDACi) such as vorinostat induce an open chromatin configuration and re-expression of epigenetically repressed genes. This can result in better chromatin access by DNA topoisomerase II (Topo II) inhibitors and/or increase the expression of Topo II. Preclinical studies using leukemia cell lines have demonstrated that the combination of vorinostat with a Topo II inhibitor has synergistic antileukemia effect (Sanchez-Gonzalez, Blood 2006). To test this concept clinically, we are conducting a CTEP sponsored clinical trial of the combination of idarubicin and vorinostat in advanced leukemia. Idarubicin was selected because it does not require age-based dose adjustment. Two different schedules of the combination are being studied. With schedule A, idarubicin 12 mg/m2 daily for 3 days is combined with vorinostat orally (PO) three times a day (TID) for 14 days (starting dose 100 mg PO TID). With schedule B, vorinostat is administered daily PO TID for only days. Cycles are repeated every 21 days. Both drugs are initiated simultaneously. Only vorinostat dose escalation is investigated, following a classic 3+3 dose escalation schema, with an expansion cohort of 10 patients at the MTD. If both schedules are open at any given time, patients are randomized among them. Thirty one patients (pts) have been treated: 15 in schedule A and 16 in B. Median age is 55 years (21–80); 28 pts (90%) had relapsed/refractory AML, 1 MDS, 1 ALL and 1 biphenotypic leukemia. 12 pts (39%) had poor risk cytogenetics and 8 (26%) had diploid cytogenetics. The median number of prior therapies was 3 (1–6). With schedule A, idarubicin at 12 mg/m2 with vorinostat at 100 mg was found to be above the MTD, with DLT’s being prolonged myelosuppression, encephalopathy, and mucositis. The expansion cohort has been completed with idarubicin at 9 mg/m2 and vorinostat at 100mg PO TID without significant toxicities. Dose escalation with schedule B continues at a dose of vorinostat at 500 mg PO TID. Grade 2 toxicities have been observed in 3 out of 3 patients at this dose. No serious cardiac toxicity has been observed on either schedule. The most common grade 1 and 2 toxicities include fatigue, nausea, diarrhea, cough, and mucositis. Thus far, 2 complete remissions and 4 complete marrow responses have been observed for an overall response rate of 23% in this refractory population. Most of these patients had failed previous anthracycline-based chemotherapy. In the samples tested thus far, histone acetylation by Western Blot and induction of p21 mRNA by RT-PCR was detected. Correlation of these biomarkers with response, as well as induction of γ-H2AX and topo II mRNA levels are being investigated. Pharmacokinetic analysis shows no significant interaction between the 2 agents and no significant differences between the 2 cohorts. There is an expected dose-dependent increase in the Cmax of vorinostat with escalating doses. Further analysis correlating serum level with response is ongoing. The combination of idarubicin and vorinostat is safe and active in AML. Histone hyperacetylation and induction of p21 is detected. Further dose escalation of the 3 day schedule and analysis of the optimal biological dose is ongoing.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2007
    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 ...
  • 7
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 2189-2189
    Abstract: Abstract 2189 Introduction: Vorinostat (Vor) is an oral HDAC inhibitor with activity in AML. In vitro, the combination of Vor with idarubicin (Ida) and ara-C (A) is synergistic. We have performed a phase I trial of Vor and Ida and demonstrated that doses of vorinostat up to 500 mg po TID daily × 3 are safe in combination with Ida. Based on this, we developed a phase II trial to study the safety and activity of the triple combination in patients (pts) with AML or higher-risk MDS. Methods: Pts ages 15 to 65 years with ECOG performance of ≤ 2 with a AML by WHO criteria or INT-2 or high risk MDS by IPSS were eligible. The study had 2 phases: an initial run-in phase followed by an actual phase II. For the run-in phase pts with relapsed or refractory disease were eligible. For the phase II, pts should not have received any prior therapy for AML or higher risk MDS. Other inclusion criteria were adequate renal, hepatic, cardiac functions. Pts with APL or CBF were excluded. In the run in phase, Vor was used at the highest possible dose (500 mg po TID × 3 days on days 1 to 3) in combination with Ida-A. Once the dose of Vor was defined in the run-in phase, the phase II was to start with a primary endpoint of event free survival (EFS). The trial was monitored every 6 months and was to be stopped early if it was unlikely that the median EFS rate will be at least (more than 2%) 7 months. The study was also monitored for Vor-related excess toxicity. Pts could receive up to 2 induction cycles, 5 cycles of consolidation and 12 of maintenance. Induction therapy: Vor 500 mg po TID × 3 days, Ida 12 mg/m2 IV over 1 hour qd × 3 (days 4 to 6) and A 1.5 g/m2 IV as a continous infusion (CI) over 24 h qd (days 4 to 7). Pts who achieve a CR or CRp (CR with incomplete platelet recovery) could receive consolidation therapy as follows: Vor 500 mg po TID × 3 days on days 1 to 3, Ida 8 mg/m2 IV over 1 hour daily × 2 days (days 4 and 5) and A 0.75 g/m2 IV CI over 24 h qd × 3 days (days 4 to 6). During maintenance phase, Vor was 200 mg po TID QD x14 every 28 days for 12 cycles. PD analysis included histone acetylation, analysis of autophagy and ROS activation. Results: 3 pts were treated in the run in phase with no toxicity. All 3 responded to therapy and the study continued to the phase II. The phase II has completed accrual at 75 pts. This was the max. number anticipated if the study was to be successful. No stopping rules for EFS or toxicity were met. Pt characteristics are as follows: median age 52 years (range 19–65), WBC 5 ku/L (range 0.7 to 111), peripheral blood blasts 14% (0-92), BM blasts 40% (11-95), cytogenetics (diploid 29 (38%), -5/-7 (22%), the rest complex), Flt3 mutation in 11 (14%). Response rate is as follows: CR 57 (76%), CRp 7 (10%) for an overall response (ORR) of 86%. Diploid pts had a CR of 86% and CRp of 7% for an ORR of 93%. Pts with other CG alterations had a CR of 76%, CRp 9%, ORR 85% (p=0.03). Pts with Flt-3 ITD had a CR of 91% and CRp of 9% for an ORR of 100%. This was in contrast to an ORR of 85% for the wt Flt-3 group (p=0.2). Four pts (6%) required 2 cycles of induction for response. Induction mortality was in 3 pts (4%). No excess toxicity was observed compared to standard Ida and Ara-C therapy. With a median follow of 6.7 (0.9 to 19.4) months: OS was 15.7 months (range 0.7 to 19.4) (64% at 1 year), remission duration (RD) 9.5 months (0-18.2) and EFS 10.2 (0.7-19.4). For diploid pts, OS was 17.7 (0.9 to 19.4), RD NR (0.7 to 14.1) and EFS NR (0.9-19.4). In pts with other CG alterations was OS was 11.7 (0.7-18.9) (p=0.3); RD 8.3 (0.7-14.1), p=0.05, and EFS 8.5 (0.7 to 15), p=0.05. Probability of survival at 1 year was 73% for diploid and 58% for others. For Flt-3+ OS was 18.2 (1.4 to 18.2) vs. 15.7 (0.9-19) in wt Flt-3 (p=0.4), RD NR (0.2 to 16.6) vs 9(0-14.5) months in wt and EFS NR vs 10.2 (0.9-15-7) in wt (p=NS). OS at 1 year was 91% for Flt-3+ vs. 60% for wt Flt-3. Induction of histone H3 acetylation on day 3 was documented in only 2 of 15 (13%) pts. Beclin, a marker of autophagy, was expressed at baseline in all 15 cases analyzed. Sequential gene expression of Nrf2, CYBB, FoxO3, SOD1,2 and GST-pi was measured sequentially by Q-PCR. No activation of any of these genes was observed with therapy. Biomarkers of DNA repair (H2AX) are ongoing. Conclusion: The combination of Ida-A Vor is safe in pts with AML/MDS. ORRs are very high with this combination, particularly in diploid and Flt-3 ITD pts. Longer follow up is needed to assess effect on survival. Studies specific for Flt3 mutated pts and in combination with standard “7+3” therapy are ongoing. 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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 30, No. 18 ( 2012-06-20), p. 2204-2210
    Abstract: To evaluate the safety and efficacy of the combination of the histone deacetylase inhibitor vorinostat with idarubicin and ara-C (cytarabine) in patients with acute myelogenous leukemia (AML) or myelodysplastic syndrome (MDS). Patients and Methods Patients with previously untreated AML or higher-risk MDS age 15 to 65 years with appropriate organ function and no core-binding factor abnormality were candidates. Induction therapy was vorinostat 500 mg orally three times a day (days 1 to 3), idarubin 12 mg/m 2 intravenously (IV) daily × 3 (days 4 to 6), and cytarabine 1.5 g/m 2 IV as a continuous infusion daily for 3 or 4 days (days 4 to 7). Patients in remission could be treated with five cycles of consolidation therapy and up to 12 months of maintenance therapy with single-agent vorinostat. The study was designed to stop early if either excess toxicity or low probability of median event-free survival (EFS) of more than 28 weeks was likely. Results After a three-patient run-in phase, 75 patients were treated. Median age was 52 years (range, 19 to 65 years), 29 patients (39%) were cytogenetically normal, and 11 (15%) had FLT-3 internal tandem duplication (ITD). No excess vorinostat-related toxicity was observed. Induction mortality was 4%. EFS was 47 weeks (range, 3 to 134 weeks), and overall survival was 82 weeks (range, 3 to 134 weeks). Overall response rate (ORR) was 85%, including 76% complete response (CR) and 9% in CR with incomplete platelet recovery. ORR was 93% in diploid patients and 100% in FLT-3 ITD patients. Levels of NRF2 and CYBB were associated with longer survival. Conclusion The combination of vorinostat with idarubicin and cytarabine is safe and active in AML.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2012
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: British Journal of Haematology, Wiley, ( 2010-04-29), p. no-no
    Type of Medium: Online Resource
    ISSN: 0007-1048 , 1365-2141
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2010
    detail.hit.zdb_id: 1475751-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 108, No. 11 ( 2006-11-16), p. 1954-1954
    Abstract: Aberrant DNA methylation and histone code alterations are common in leukemia. The oral isotype-selective HDAC inhibitor MGCD0103 and the DNA methyltransferase inhibitor azacitidine have been shown to have synergistic antileukemia activity in preclinical models. Both agents have single-agent clinical activity in MDS and AML (Garcia-Manero, ASCO, 2006 & Silverman, JCO, 2002). Based on this, we have developed a Phase I/II study of the combination in leukemia. Patients with relapsed/refractory MDS (10% or more marrow blasts) or AML, or untreated older patients with AML are eligible. Adequate performance status, renal and hepatic functions are required. Azacitidine is administered at its approved dose/schedule: 75 mg/m2 SC daily x 7 every 28 days. MGCD0103 was started on day 5 of azacitidine and was given as an oral dose 3 times a week without adjusting for body weight, without interruption. The phase I portion of the study design followed a classic “3+3” model and only MGCD0103 was dose escalated. Three dose levels of MGCD0103 have been studied so far: 35, 60 and 90 mg. Twelve patients have been registered in this study. The median age is 61 (range 45–85). All, but 1 patient with MDS, had AML. All patients had relapsed or refractory disease. A total of 24 cycles have been administered, mean = 2.0 (range 1–4). Only 1 patient has experienced dose limiting toxicity: grade 3 vomiting at a dose of 90 mg of MGCD0103. Otherwise, the combination has been very well tolerated and the MTD has not yet been defined, with dosing currently ongoing at the approximate MTD of single agent MGCD0103: 110 mg orally three times a week without interruption. Preliminary PK data indicate that the t1/2 for azacitidine is less than 2 h, and does not appear to be affected by MGCD0103. Likewise, MGCD0103 pharmacokinetic characteristics do not appear to be affected by azacitidine. The majority of patients exhibited substantial reduction in HDAC activity during treatment with the combination. Analysis of DNA methylation is ongoing. Two patients have achieved response: 1 complete remission after 4 courses of therapy (in an older patient in first relapsed AML with an initial CR duration of 11 months and multiple cytogenetic abnormalities). A second older patient with refractory AML achieved a complete marrow CR (marrow blasts less than 5%) but died from pneumonia (not drug-related) after the second course of therapy on day 28 before peripheral count recovery. In conclusion, the combination of azacitidine with MGCD0103 is safe in patients with advanced AML/MDS. The combination has encouraging safety, PK, and clinical activity profiles. The study continues at the MTD of single agent MGCD013 in combination with azacitidine. Once the MTD of the combination is documented, the study will continue as a phase II study in this patient population.
    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
    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