Phase I/II Study of BI 2536, An Intravenous Polo-Like Kinase-1 (Plk-1) Inhibitor, in Elderly Patients with Relapsed or Refractory Acute Myeloid Leukemia (AML): First Results of a Multi-Center Trial

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2973-2973
Author(s):  
C. Müller-Tidow ◽  
Gesine Bug ◽  
Richard Schlenk ◽  
Michael Lübbert ◽  
Alwin Krämer ◽  
...  

Abstract Elderly patients with refractory or relapsed AML have a dismal prognosis and new treatments are needed for this patient population. BI 2536 is a potent and selective inhibitor of Plk-1, which plays a crucial role in the regulation of mitosis. BI 2536 demonstrated strong anti-proliferative effects on AML cells in vitro and in vivo xenograft models and is the first specific Plk-1 inhibitor in clinical testing. Here, we present preliminary results of a Phase I/II study of single-agent BI 2536 therapy in patients 60 years of age or older with relapsed or refractory AML. Three administration schedules were tested: BI 2536 was given as an intravenous 1h-infusion on day 1 (schedule A), on days 1, 2, and 3 (schedule B), or on days 1 and 8 (schedule C) in 3-week cycles, respectively. In the phase I part, the 3+3 dose escalation design was used to evaluate the maximum tolerated dose (MTD) in AML patients. Dose escalation started at the respective MTD as previously determined in solid tumor patients. The phase II part consisted of extension cohorts at the MTD to further investigate safety and efficacy. The following dose levels of BI 2536 were evaluated: 200, 250, 300, 350, and 400 mg in schedule A; 50 and 60 mg in schedule B; 100, 150, 200, and 225 mg in schedule C. Initially, only schedule A and B were tested. As the blast reduction was only of short duration, we decided to discontinue schedule B after the 60 mg dose level and to introduce schedule C to shorten the interval between drug administrations. Currently, 60 evaluable patients (33 males, 27 females) have been treated: median age 68.5 years (range 61 – 82); ECOG score from 59 patients: 0: 10, 1: 32, 2: 17; secondary AML in 31 (53%) of 58 patients; complex karyotype in 7 (14%) of 49 patients; median number of previous therapies = 4 (range 1 – 13); in 50 patients, data on best response to previous therapies were available: CR = 25, PR = 4, no change = 17, PD = 4. In total, 119 cycles of trial treatment were administered in 60 patients (median = 1, range 1 – 7). The most frequent drug-related AEs were thrombocytopenia (21%), alopecia, anemia, and neutropenia (each 18%), nausea (15%), constipation, cough, fatigue, infection, leukopenia, and mucosal inflammation (each 12%). Dose limiting toxicities occurred in 8 patients and included neutropenic fever (3 cases), sepsis, pneumonia/sepsis, intracranial bleeding/hyphema, somnolence/coma and anal thrombosis. The MTD was determined at 350 mg in schedule A and at 200 mg in schedule C. No MTD was determined for schedule B. A greater than 50% reduction of blasts in the peripheral blood was seen in 17 of 36 evaluable patients after treatment with BI 2536; however, the effect on blast counts was only transient in most patients. Of 58 patients evaluable for response, one patient achieved a CR, one a CRi, and one a PR. Twenty-one patients had temporarily stable blood values, 31 progressed after the first cycle. In 3 patients, the response was indeterminate. The 24 patients who achieved remission or did not progress after the first cycle received a median of 3 treatment cycles (range 1 – 7). Dose-dependent pharmacodynamic activity was demonstrated by flowcytometric and immunocytochemical analysis of the bone marrow before and after BI 2536 administration. Pharmacokinetic data will be reported at the meeting. In conclusion, BI 2536 as single agent therapy is well tolerated and can be administered at higher doses in relapsed or refractory AML than in solid tumors. BI 2536 demonstrated clinical activity in patients with relapsed and treatment refractory AML.

2019 ◽  
Vol 37 (15_suppl) ◽  
pp. 3059-3059 ◽  
Author(s):  
Junning Cao ◽  
Pin Zhang ◽  
Paul L. de Souza ◽  
Bo Gao ◽  
Mark Voskoboynik ◽  
...  

3059 Background: Poly (ADP-ribose) polymerase (PARP) enzymes play critical roles in DNA damage detection and repair. IMP4297 is a novel, potent PARP1/2 inhibitor (IC50 6.27nM/1.57nM) and has demonstrated to be 20-fold more potent than Olaparib in anticancer animal models. Two phase I studies were performed to evaluate and characterize the tolerability and safety, pharmacokinetics, and antitumor activity of single agent IMP4297 in Chinese and Australian patients with advanced ovarian, breast, prostate and other solid tumors. Methods: Dose escalation used a 3+3 design with a modified Fibonacci escalation. Dose cohort expansion was planned after efficacy was observed at the lowest dose level. Patients received IMP4297 monotherapy orally once a day until disease progression or unacceptable toxicity. Results: As of Jan 12, 2019, 56 patients, including 23 BRCA mutation carriers (BRCA+), had been enrolled at 2-100 mg dose level. No DLT was observed. In these two studies, the most frequent treatment-related adverse events (TRAEs) were leukopenia (20%), followed by anemia (18%), nausea (18%) and thrombocytopenia (14%). The majority of TRAEs were grade 1 or 2. Grade 3 TRAEs occurred in five patients (anemia, n=2; vomiting, n=1; thrombocytopenia, n=1; elevated AST, n=1). Only one patient had a dose reduction due to grade 3 thrombocytopenia. No serious TRAEs were observed. In 15 BRCA+ patients who had measurable lesions, the ORR was 33% and the DCR was 80%. There were 4 BRCA+, platinum-sensitive ovarian cancer patients with an ORR of 75% and a DCR of 100%. One patient with somatic BRCA mutated urothelial carcinoma showed a 76% decrease in tumor size. Conclusions: IMP4297 has been well-tolerated with significant anti-tumor activity. The 100 mg daily dose was selected as the RP2D based on safety, pharmacokinetics and clinical activity, and will be further characterized in dose expansion and phase II studies. Tumor response to treatment (RECIST 1.1) in patients with measurable lesions. Clinical trial information: NCT03508011 and NCT03507543. [Table: see text]


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 3113-3113
Author(s):  
Seock-Ah Im ◽  
SeungHwan Lee ◽  
Keun Wook Lee ◽  
Youngjoo Lee ◽  
Joohyuk Sohn ◽  
...  

3113 Background: JPI-547 is an oral inhibitor of PARP 1/2 and Tankyrase 1/2. JPI-547 demonstrated anti-tumor activity in BRCA-deficient xenograft models as a single-agent and in combination with chemotherapy and immune checkpoint inhibitors. Methods: This is the first in human (FIH) phase I study of JPI-547 in patients (pts) with advanced solid tumors. For the dose escalation phase, a 3+3 design was used with 4 doses from 50 to 200 mg QD on 21-day cycles. Primary objectives were to assess safety and tolerability to determine RP2D, and secondary objectives included pharmacokinetics and preliminary antitumor activities. DLT monitoring period was 21 days. Pharmacodynamics and information of HRR mutation were also explored. For the dose expansion phase, pts with documented pathogenic germline or somatic BRCA/HRR mutations were enrolled to assess the preliminary efficacy and safety. Tumor response (RECIST 1.1) was evaluated every 6 weeks. Centralized germline BRCA testing was conducted to confirm pathogenic gBRCA mutations. Results available at the cut-off date of 31-Dec-2020 are presented. Results: For dose escalation phase, 22 pts were enrolled. JPI-547 was well absorbed with Tmax of 0.25-8 h post-dose and apparent half-life of 18-31 h. Mean Cmax and AUC increased proportionally (within the dose range of 50-200 mg). PAR level measured from PBMC was 53% inhibited at Cmax. One DLTs was observed at 100 mg (elevated ALT, G3) and 200 mg (elevated ALT/AST, G3) respectively. MTD was determined as 200 mg after considering DLTs and myelosuppression observed from cycle 2. RP2D was determined to be 150 mg based on the pharmacokinetic data and safety. Thirteen pts (59.1%) had at least one grade 3/4 TRAE and 12 had dose interruption/reduction due to TRAE. The most common ( > 20%) TRAE were anemia, thrombocytopenia and neutropenia. In dose expansion phase, 40 pts were enrolled, and response was evaluable in 39 pts. The best overall responses were 11 confirmed PR (cPR) and 15 SD with ORR of 28.2% (11/39) and DCR of 64.1 % (25/39). The mPFS was 3.5 mos and mDoR was 3.4 mos. At the time of data cut-off, three pts were ongoing as following response and cancer types: cPR (breast, ATMm, 9.0 mos), cPR (NSCLC, gBRCA2m, 3.8 mos) and SD (breast, gBRCAm, 9.3 mos). Five pts (2 ovarian, 3 breast) previously treated with olaparib and discontinued due to progressive disease were enrolled in this JPI-547 trial and one ovarian cancer pt showed cPR with 37% tumor shrinkage. Conclusions: These results demonstrate that JPI-547 is adequately absorbed with acceptable safety profile. Preliminary efficacy results suggest that JPI-547 monotherapy is effective in pts with BRCA/HRR mutation. Further investigation is warranted in pts with solid tumor including PARP inhibitor resistant cases. Clinical trial information: NCT04335604.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3316-3316 ◽  
Author(s):  
Gesine Bug ◽  
Richard F. Schlenk ◽  
Carsten Müller-Tidow ◽  
Michael Lübbert ◽  
Alwin Krämer ◽  
...  

Abstract Abstract 3316 Background: Patients with refractory or relapsed AML have a poor prognosis and new treatments are needed for this patient population. While younger AML patients might benefit from intensive salvage treatments, a substantial number of elderly patients are considered ineligible for intensive treatment approaches. For these patients, repeated cycles of low-dose cytarabine (LD-Ara-C) are an accepted therapeutic option for palliative treatment. The serine/threonine kinase Polo-like kinase 1 (Plk1) controls several key steps in mitosis. BI 6727 is a first in class, highly selective and potent cell cycle kinase inhibitor targeting Plk1, and has demonstrated antiproliferative activity in multiple cell lines and animal models. Targeting Plk1 with BI 6727 results in cell cycle arrest in prometaphase (referred to as polo arrest) leading to eventual apoptosis. In a phase I dose escalation trial in patients with advanced solid tumors a favorable safety profile and encouraging antitumor activity was reported. BI 6727 has demonstrated a long terminal half life of 111 hours and a high volume of distribution suggesting excellent tissue distribution in patients. Here, we present preliminary results from the Phase I part of an ongoing Phase I/II study of BI 6727 in combination with LD-Ara-C in patients with relapsed or refractory AML considered ineligible for intensive treatment. Methods: This study follows a two stage design: the maximum tolerated dose (MTD) of BI 6727 in combination with fixed dose LD-Ara-C was evaluated in the Phase I dose escalation part of the trial following a 3+3 design with de-escalation. In a second ongoing treatment schedule the MTD of single agent BI 6727 is investigated, the MTD of single agent BI 6727 has not been reached yet. In the planned randomized Phase II part of the study, efficacy of BI 6727 plus LD-Ara-C will be compared to LD-Ara-C alone. BI 6727 was administered as a one hour intravenous infusion on days 1+15 every 28 days in combination with fixed dose LD-Ara-C (20 mg bid s.c). The BI 6727 starting dose was based on the MTD previously determined in solid tumor patients. Patients with no progression after the first cycle were allowed to continue treatment. Results: Patient characteristics were as follows: median age was 71 years (range 40 – 81); ECOG performance score 0: 9 pts; 1: 17 pts; 2: 5 pts. Increasing BI 6727 doses in combination with LD-Ara-C were evaluated in 31 patients (21 males, 10 females). Safety: Drug related adverse events (AEs) were reported in 17 of the 31 patients. The most frequent AEs reported (>5%) were: anemia and febrile neutropenia (each 9.7%), infections (pneumonia), decreased appetite and headache (each 6.5%). Dose-limiting toxicities (DLTs) were reported in 4 patients treated with BI 6727 + LD-Ara-C. DLTs as rated per protocol were: pneumonia, mucositis, hypersensitivity/allergic reaction and myocardial infarction. Based on the preliminary reports on DLTs the MTD for BI 6727 in combination with LD-Ara-C was determined. Preliminary response data of 28 patients with relapsed/refractory AML treated at different BI 6727 doses in combination with LD-Ara-C are available: 5 patients achieved a CRi or CR, 2 patients achieved a PR. Six patients had temporarily stable blood values (“no change” as best response). 10 patients suffered from progression during or at the end of the 1st treatment cycle, and 5 patients were ineligible for response assessment. An update of the phase I part of this trial with further details on patient/disease characteristics, safety and efficacy of BI 6727 in combination with LD-Ara-C will be reported at the meeting. Conclusion: Preliminary results indicate that BI 6727 in combination with LD-Ara-C is well tolerated in patients with relapsed/refractory AML ineligible for intensive treatment. The MTD of BI 6727 in combination with LD-Ara-C was determined. BI 6727 in combination with LD-Ara-C showed first signs of clinical activity in AML patients. Safety and efficacy of BI 6727 + LD-Ara-C will be further explored in the phase II part of the trial. Disclosures: Off Label Use: LD-Ara-C in combination with BI 6727 for treatment of patients with relapsed refractory AML ineligible for intensive treatment. Fleischer:Boehringer Ingelheim Pharma GmbH & Co KG: Employment. Taube:Boehringer Ingelheim Pharma GmbH & Co KG: Employment.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2605-2605
Author(s):  
Sameem Abedin ◽  
Guru Subramanian Guru Murthy ◽  
Lyndsey Runaas ◽  
Laura C. Michaelis ◽  
Ehab L. Atallah ◽  
...  

Background: Lintuzumab Ac225 is a radiolabeled anti-CD33 antibody with demonstrated single-agent activity in AML. The drug is composed of an alpha emitting isotope, Ac225, conjugated with the humanized anti-CD33 monoclonal antibody lintuzumab. In a prior study, lintuzumab Ac225 monotherapy was safely administered to elderly/unfit AML patients with toxicities primarily limited to prolonged myelosuppression. Nearly 70% of patients achieved a CR/CRi at the highest dose level (2uCi/kg). We hypothesized that lintuzumab Ac225, when added to the salvage chemotherapy regimen CLAG-M (cladribine, cytarabine, G-CSF, and mitoxantrone), would be tolerable and would improve remission rates in the treatment of relapsed/refractory AML (RR-AML). CLAG-M salvage was selected based on favorable institutional outcomes (CR/CRi: 54%, Mushtaq et al, ASH 2018). This novel investigator-initiated phase I study is the first study to combine radioimmunotherapy and intensive chemotherapy in patients with RR-AML. Patients and Methods: Eligible patients include medically fit, RR-AML patients aged 18 years and older, with adequate organ function. In addition, more than 25% of leukemic blasts must have been CD33 positive by flow cytometry. Induction consisted of G-CSF, 300mcg/d, given D1-6, cladribine 5mg/m2, given D2-6, cytarabine 2g/m2, given D2-6, and mitoxantrone 10mg/m2, given D2-4. Lintuzumab Ac225 was administered as a single dose on either days 7, 8, or 9. Cohort 1 received lintuzumab Ac225 at a dose of 0.25uCi/kg, and cohort 2 received a dose of 0.50uCi/kg. Only one induction course was administered, and subsequent treatments were up to physician discretion. Results: Nine patients with a median age of 59 years (range 47-73 yrs) have been enrolled. Patients had a median of 2 (range 1-4) prior, anti-leukemic treatments, including 4 patients who relapsed following allogeneic HCT. Four patients (44%) had intermediate risk cytogenetics, and five patients (56%) had adverse risk features. Median blast CD33 expression was 73% (range 32-100%). All patients completed one cycle of lintuzumab Ac225 with CLAG-M. Three patients enrolled into cohort 1 (0.25mcg/kg), and six patients enrolled into cohort 2 (0.5mcg/kg). Among all enrolled patients, Grade 3 or greater AEs include febrile neutropenia (n=4), infection (n=4), QTc prolongation (n=1), hypophosphatemia (n=1), hyponatremia (n=1), and tumor lysis syndrome (n=1). In two responding patients enrolled to cohort 2, one patient had prolonged time to neutrophil recovery (ANC>500 at 53 days), and a second patient, who was post-HCT, had an ANC recovery to 300 by day 42, and then received donor CD34+ stem cells to aide in count recovery. Figure 1 summarizes toxicities. All responding patients with a platelet count>50k prior to therapy, had a post-treatment platelet count >50k within 42 days. No mortalities were observed on study. Among cohort 1 patients (n=3), one patient achieved CR, one patient had a >50% blast reduction to 6.2% blasts with neutrophil recovery and platelet improvement to 70k, and the third patient had no response. Overall, 33% (1/3) achieved a CR. Among patients enrolled into cohort 2, two patients achieved CR, two patients achieved CRp, one patient achieved CRi, and one patient had no response. Overall, 83% (5/6), achieved CR/CRp/CRi. After, three patients on study subsequently went onto allogeneic HCT, two patients initiated donor lymphocyte infusions (DLI), and one patient underwent CD34+ stem cell boost. Conclusion: Combining lower doses of lintuzumab Ac225 with salvage CLAG-M chemotherapy appears to have a clinically acceptable safety profile. With dose escalation, increased myelosuppression has been noted alongside, but also highly encouraging efficacy results for RR-AML. As two patients in Cohort 2 met DLT criteria due to prolonged ANC recovery, this study will be amended to mandate the addition of G-CSF two weeks after Lintuzumab Ac225 treatment. Tentatively, this study will plan to extend the ANC recovery observation period to 60 days. Depending on safety and further efficacy data, this may lead to further dose escalation, or potentially an expanded number of patients in a Phase 2 study. Disclosures Abedin: Actinium Pharmaceuticals: Research Funding; Jazz Pharmaceuticals: Honoraria; Agios: Honoraria; Helsinn Healthcare: Research Funding; Pfizer Inc: Research Funding. Runaas:Agios: Honoraria; Blueprint Medicine: Honoraria. Michaelis:Incyte: Consultancy, Research Funding; Novartis: Consultancy; BMS: Research Funding; TG Therapeutics: Consultancy, Research Funding; JAZZ: Other: Data Safety Monitoring Board, uncompensated, Research Funding; Macrogeneics: Research Funding; Millenium: Research Funding; ASTEX: Research Funding; Pfizer: Equity Ownership, Research Funding; Celgene: Consultancy, Research Funding; Bioline: Research Funding; Janssen: Research Funding. Atallah:Helsinn: Consultancy; Jazz: Consultancy; Takeda: Consultancy, Research Funding; Pfizer: Consultancy; Jazz: Consultancy; Helsinn: Consultancy; Novartis: Consultancy. Hamadani:Celgene: Consultancy; Merck: Research Funding; Sanofi Genzyme: Research Funding, Speakers Bureau; Otsuka: Research Funding; Janssen: Consultancy; Medimmune: Consultancy, Research Funding; ADC Therapeutics: Consultancy, Research Funding; Takeda: Research Funding; Pharmacyclics: Consultancy.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. 3640-3640
Author(s):  
Filip Janku ◽  
Elena Elez ◽  
Gopa Iyer ◽  
Noboru Yamamoto ◽  
Daniel Shao-Weng Tan ◽  
...  

3640 Background: LTT462 is an investigational small molecule inhibitor of ERK1/2, which has demonstrated preclinical activity in multiple MAPK activated cancer cells and xenograft models. This first-in-human study was designed to evaluate the safety and tolerability of LTT462 in advanced solid tumors harboring MAPK pathway alterations (NCT02711345). Methods: The dose-escalation part of this Phase I, open-label study, enrolled adult and adolescent pts with advanced solid tumors harboring ≥1 documented MAPK pathway alteration with progressive disease (PD) despite standard therapy, or for whom there is no effective standard treatment. Oral LTT462 was given once daily (QD) at 45–600 mg or twice daily (BID) at 150 mg or 200 mg. Objectives were to determine the maximum tolerated dose (MTD) using a Bayesian hierarchical logistic regression model guided by escalation with overdose control, and characterize safety, pharmacokinetics, pharmacodynamics, and preliminary efficacy of LTT462. Results: Sixty-five pts (median age 60 years) including 1 pt aged 15 were enrolled in the dose-escalation; most pts (22%) had 3 prior therapies. Most common primary sites for cancer were in the colon (n = 21; 32%), ovary (n = 9; 14%), and pancreas (n = 7; 11%). All pts discontinued, the majority due to PD (n = 44; 68%). Eleven pts experienced DLTs; 6 pts experienced Grade 3 eye disorder DLTs (4 pts retinopathy, 2 pts chorioretinopathy). Treatment-related adverse events (TRAEs) were reported for 89% of pts, most commonly ( > 30%) diarrhea (n = 25; 38%) and nausea (n = 22; 34%). Grade 3/4 TRAEs were reported in 29% of pts; most common was retinopathy (n = 4; 6%). MTD of LTT462 was 400 mg QD and 150 mg BID. Overall, 8 pts (12%) had stable disease (SD) and 35 pts (54%) had PD. An unconfirmed partial response was reported in a pt with cholangiocarcinoma with BRAF mutation; best change in sum of target lesions per RECIST 1.1 was -33.9%. LTT462 increased plasma peak drug concentration and drug exposure at increasing doses between 45–450 mg QD. Exposure at LTT462 600 mg QD was lower than anticipated, indicating potential saturation of absorption at this dose. LTT462 inhibited ERK1/2 and reduced DUSP6 expression relative to baseline in most pts evaluated. Conclusions: LTT462 is well tolerated. Limited clinical activity was reported with single agent LTT462; best overall response was SD. An ongoing study is investigating LTT462 in combination with the RAF inhibitor, LXH254, in NSCLC and melanoma. Clinical trial information: NCT02711345 .


2020 ◽  
Vol 38 (21) ◽  
pp. 2418-2426 ◽  
Author(s):  
Daniel V.T. Catenacci ◽  
Drew Rasco ◽  
Jeeyun Lee ◽  
Sun Young Rha ◽  
Keun-Wook Lee ◽  
...  

PURPOSE To evaluate the safety, pharmacokinetics, and preliminary activity of bemarituzumab in patients with FGFR2b-overexpressing gastric and gastroesophageal junction adenocarcinoma (GEA). PATIENTS AND METHODS FPA144-001 was a phase I, open-label, multicenter trial consisting of the following 3 parts: part 1a involved dose escalation in patients with recurrent solid tumors at doses ranging from 0.3 to 15 mg/kg; part 1b involved dose escalation in patients with advanced-stage GEA; and part 2 involved dose expansion in patients with advanced-stage GEA that overexpressed FGFR2b at various levels (4 cohorts; high, medium, low, and no FGFR2b overexpression) and 1 cohort of patients with FGFR2b-overexpressing advanced-stage bladder cancer. RESULTS Seventy-nine patients were enrolled; 19 were enrolled in part 1a, 8 in part 1b, and 52 in part 2. No dose-limiting toxicities were reported, and the recommended dose was identified as 15 mg/kg every 2 weeks based on safety, tolerability, pharmacokinetic parameters, and clinical activity. The most frequent treatment-related adverse events (TRAEs) were fatigue (17.7%), nausea (11.4%), and dry eye (10.1%). Grade 3 TRAEs included nausea (2 patients) and anemia, neutropenia, increased AST, increased alkaline phosphatase, vomiting, and an infusion reaction (1 patient each). Three (10.7%) of 28 patients assigned to a cohort receiving a dose of ≥ 10 mg/kg every 2 weeks for ≥ 70 days reported reversible grade 2 corneal TRAEs. No TRAEs of grade ≥ 4 were reported. Five (17.9%; 95% CI, 6.1% to 36.9%) of 28 patients with high FGFR2b-overexpressing GEA had a confirmed partial response. CONCLUSION Overall, bemarituzumab seems to be well tolerated and demonstrated single-agent activity as late-line therapy in patients with advanced-stage GEA. Bemarituzumab is currently being evaluated in combination with chemotherapy in a phase III trial as front-line therapy for patients with high FGFR2b-overexpressing advanced-stage GEA.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1953-1953 ◽  
Author(s):  
Richard Schlenk ◽  
Kristina Sohlbach ◽  
Marie-Luise Hütter ◽  
Patrice Ceballos ◽  
Nathalie Fegueux ◽  
...  

Abstract Patients either with refractory/relapsed AML or with newly diagnosed AML not suitable for intensive induction therapy have a dismal prognosis. Belinostat belongs to a new class of hydroxymate-type histone deacetylase (HDAC) inhibitors. Belinostat has demonstrated effective cell killing in leukemic and lymphoma cells as a single agent and synergy in combination with other therapeutic regimens such as anthracyclines. Preliminary results are presented of the still ongoing open-label non-randomized, multi-centre, phase I/II trial to assess the efficacy and safety of 2 schedules of belinostat in combination with idarubicin in patients with AML not suitable for standard intensive therapy. Belinostat was administered either as a daily 30 min infusion for 5 days q 3 weeks plus iv idarubicin on days 4+5 (regimen A) or as a continuous iv administration over 48 hours q 2 weeks with addition of iv idarubicin at h 24 and 48 (regimen B). Both schedules used a dose escalation design; regimen A, the 3+3 dose escalation design; regimen B, the accelerated titration design. Currently, 22 patients have been treated (regimen A, n=11; regimen B, n=11). Patient characteristics at inclusion were as follows: median age 72 years (range 37–84); 12 had de novo AML, all were in relapse from previous treatment, 4 had secondary AML, two had received prior treatment, 7 had MDS/AML, 3 received prior treatment. In total, 49 cycles of trial treatment were administered in 22 patients (median = 2, range 1 – 8). The safety database as of 11.07. 2008 with information available for 19 patients (n=10 in regimen A. n=9 in regimen B) only contained two SAE’s defined by the investigators as related to treatment, they occurred in the same patient and were of the same type neutropenia and thrombocytopenia, conditions known as associated with idarubicin and also fundamentally a part of the progressive leukemic process and therefore in part exempted from the DLT definition in this as in other AML treatment protocols. So far no dose limiting toxicity was defined. Most frequent related adverse events, occurring in less than one third of the patients, were nausea, vomiting, diarrhea. Antileukemic efficacy was seen both following belinostat single agent (CRi after 2 cycles in one 62 yr old MDS/AML patient receiving 48h CIV at a dose of 800 mg/m2/d, regimen B) and following belinostat in combination with idarubicin (CRi after one cycle in one 84 yr old MDS/AML patient receiving 48h CIV at a dose of 1000 mg/m2/d plus idarubicin 5 mg/m2 x1, regimen B; CR after one cycle in one 74 yr old AML patient receiving the 5-d regimen of belinostat 1000 mg/m2/d + idarubicin 10 mg/m2 x 1 and extramedullary cutaneous relapse after the 8th cycle, regimen A; CRi after 3 cycles in one 78 yr old MDS/AML patient receiving the 5-d regimen of belinostat 1000 mg/m2/d + idarubicin 7.5 mg/m2 x 2, regimen A). Updated results and pharmacokinetic analysis will be presented. Conclusion: In the ongoing PXD101-CLN-15 study no dose limiting toxicity was determined so far. Clinical efficacy in the form of complete remissions has been demonstrated both of the belinostat monotherapy and of the combination with idarubicin in the 5-d regimen as well as in the CIV regimen. Dose escalation continues as preparation for a phase 2 expansion.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2065-2065
Author(s):  
William Blum ◽  
Rebecca B. Klisovic ◽  
Alison Walker ◽  
Ramiro Garzon ◽  
Shujun Liu ◽  
...  

Abstract Abstract 2065 Poster Board II-42 Background: Hypomethylating agents have significant clinical activity in myelodysplastic syndromes (MDS) and AML. In AML, we recently demonstrated a novel epigenetic mechanism of action for the proteasome inhibitor bortezomib (Liu, Blood 2008). Bortezomib induced hypomethylation of leukemic cells in vitro and in vivo via depletion of the Sp1/NF-kB transcriptional activation complex on the DNA methyltransferase 1 (DNMT1) gene promoter, which results in down-regulation of DNMT1 mRNA and enzyme, DNA hypomethylation and re-expression of otherwise hypermethylated target genes. Based on this preclinical work, we designed a phase I dose escalation study of 5-azacytidine (AZA) in combination with bortezomib in AML. Methods: Adults with relapsed or refractory AML by WHO criteria and preserved organ function with ECOG ≤2 were eligible. Previous decitabine or AZA was permitted. Patients received AZA at 75mg/m2 IV daily for days (d) 1-7. Bortezomib was gradually dose escalated–dose level 1 (DL 1): 0.7mg/m2 by IV push given immediately after AZA on d 2 and 5; DL 2: 0.7mg/m2 on d 2, 5, 9, and 12; DL 3: 1.0mg/m2 on d 2, 5, 9, and 12; and DL 4: 1.3mg/m2 on d 2, 5, 9, and 12. Cycles were repeated every 28 d, regardless of count recovery or response at least until 3 cycles were administered. Responses were graded by International Working Group criteria for AML (Cheson, JCO 2003). Bortezomib was discontinued after 3 cycles if no objective response of complete remission (CR), CR with incomplete count recovery (CRi), or partial remission (PR) was achieved, but AZA could be continued beyond this timepoint in the absence of disease progression. For responding patients, 12 or more cycles of therapy were permitted. Dose limiting toxicities (DLT) were assigned for cycle 1 of therapy. Given the high likelihood of infection in this population regardless of therapy, infection was not considered a DLT. Six additional patients were treated at the recommended phase 2 dose (RP2D). Results: 23 patients were enrolled with a median age of 65 years (range, 42-81) and had received a median of 2 prior inductions (range, 1-5). Median presenting WBC was 3,700/uL (500-59,100/uL); median BM blast was 26% (2-93%). 14 patients were refractory to last therapy received, including 4 with primary refractory AML. 9 patients had relapsed disease; all but 2 of these had prior CR duration <1 year. Patients received a median of 2 cycles of study therapy (range, 1-12+ cycles). Dose escalation was halted once the target bortezomib dose was reached; the RP2D was AZA at 75mg/m2 d 1-7 plus bortezomib 1.3mg/m2 d, 2, 5, 9, 12. Though no toxicities were considered to be DLT in this study, infection and/or febrile neutropenia were universal. Death within 8 weeks occurred in 5 patients (22%) due to pneumonia (1), sepsis (1), or progressive disease (3). Two patients had discontinuation of bortezomib after 2 cycles due to Grade 3 neuropathy; only 1 patient received bortezomib beyond 3 cycles. In 3 patients without objective response (and with no progression), AZA alone was continued after 3 cycles of combination therapy; each reported a subjective improvement in fatigue without bortezomib. Overall, the objective response rate was 26% (6/23). Responses were as follows: 3- CR, 2- CRi, and 1-PR. One CRi patient (in cytogenetic remission also) who discontinued study treatment after 2 cycles due to unrelated trauma subsequently had complete count recovery, but a repeat marrow examination was not performed. Three patients went on to allogeneic transplantation due to response achieved. Response followed the typical pattern of azanucleoside activity, requiring more than one cycle of therapy; the median number of cycles to initial response was 2 (range, 1-5). 5/6 responders had response to combination therapy; one patient responded following 5 cycles of treatment, the last 2 cycles with AZA as a single agent. Conclusions: The combination of 5-azacytidine and bortezomib is well tolerated and active in this cohort of relapsed or refractory AML patients. Additional studies to further elucidate the role of proteasome inhibition as a mediator of hypomethylating activity in AML are warranted. Correlatives studies are ongoing. Disclosures: Blum: Celgene: Research Funding.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 2078-2078 ◽  
Author(s):  
Y. Ko ◽  
S. North ◽  
S. R. Berry ◽  
D. S. Ernst ◽  
L. Klotz ◽  
...  

2078 Background: GTI-2501 (GTI) is a 20-mer oligonucleotide that is complementary to the R1 subunit of the RNR mRNA. The R1 protein is overexpressed in multiple tumour cell lines. GTI displays anti-tumour activity against prostate cancer xenografts in mice as a single agent and in combination with mitoxantrone. GTI also adds to the anti-tumour efficacy of taxanes in breast cancer xenografts in mice. A Phase 1 study of a 14 day continuous infusion of GTI in patients with solid tumours showed no dose limiting toxicities at doses up to 210.9 mg/m2/day. The objective of this dose escalation phase of a phase I/II study was to define a safe phase II dose of GTI in combination with docetaxel (D) in men with metastatic hormone refractory prostate cancer (HRPC). Methods: Men with metastatic HRPC were enrolled at 3 centres in Canada. GTI was given as a 14d continuous IV infusion every 21d with D IV infusion started 2 hrs prior to the end of the GTI infusion. Planned dose escalation cohorts are summarized in table . Results: 13 men were enrolled to the 3 cohorts. All patients are evaluable for toxicity. There was one possible DLT - an episode of grade 4 neutropenia reported at cycle 2 day 1 in the highest dose cohort - but the duration of neutropenia could not be confirmed. 3 additional patients were accrued to that cohort with no DLTs. The most common gr 3/4 toxicity was attributable to D (10 pts with Gr 3/4 neutropenia). The observed incidence of Gr 3/4 neutropenia was expected since patients had weekly CBCs. Only 1 patient had febrile neutropenia. 11 pts had fatigue (4 Gr 3) related to D and /or GTI. Other GTI attributable adverse events were Gr 1/2 including transient rises in transaminases and PTT. The pharmacokinetic data which is summarized in the table will be presented in full at the meeting. Conclusions: GTI can be given safely at its highest planned dose with standard doses of D. A Phase II evaluation of the GTI + D combination is planned for men with HRPC. [Table: see text] [Table: see text]


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 3023-3023 ◽  
Author(s):  
Carlos Becerra ◽  
Jeffrey R. Infante ◽  
Lawrence E. Garbo ◽  
Michael S. Gordon ◽  
David A. Smith ◽  
...  

3023 Background: Trametinib, an oral MEK1/MEK2 inhibitor, has demonstrated single-agent clinical activity. In vitro studies of trametinib plus docetaxel (doc), pemetrexed (pem) and erlotinib (erl) showed enhanced growth inhibition of lung cancer cell lines with and without RAS/RAF mutations. Trametinib+doc significantly increased apoptosis compared with either agent alone. Methods: This is a two-part, five-arm, phase I/Ib, open-label study to evaluate the safety and tolerability of trametinib plus doc, erl, pem, pem+carboplatin (pem+carbo), or nab-paclitaxel (nab-pac) (NCT01192165). Part I is dose escalation in patients (pts) with advanced solid tumors; part II is dose expansion in pts with lung and pancreatic cancers. A 3+3 dose-escalation design was used to determine the maximum tolerated dose (MTD) and the recommended phase II regimen (RP2R) for each combination. Dose-limiting toxicities (DLTs) were determined during the first treatment cycle (21 days). Trametinib was started at 0.5 mg/day; chemotherapy was given at full recommended doses. Erl was escalated from 50 mg/day. Pharmacokinetic (PK) samples were collected pre-, and 1, 2, 3 and 6 hours post-dose. Results: As of January 2012, 80 pts have been enrolled across all arms except trametinib+nab-pac. Preliminary exposure results of trametinib+doc, erl, or pem suggest no PK drug-drug interaction. The predominant DLT for trametinib+doc without growth factors (MTD = 0.5 mg+60 mg/m2) was neutropenia. When administered with growth factors, trametinib+doc has been given up to 1.5 mg+75 mg/m2 with no DLTs. The predominant DLTs for trametinib+erl (MTD = 1 mg+100 mg) were diarrhea and mucositis and for trametinib+pem (MTD = 1.5 mg+500 mg/m2) were mucositis and febrile neutropenia. The MTD for trametinib+pem+carbo has not yet been determined. To date there are 5 PRs in the trametinib+doc group and 2 PRs in the trametinib+pem group. An NSCLC expansion cohort for trametinib+pem is enrolling. Conclusions: Trametinib+doc and trametinib+pem have shown acceptable tolerability and initial evidence of clinical activity.


Sign in / Sign up

Export Citation Format

Share Document