scholarly journals Interim Data from a Randomized, Placebo Controlled, Phase 1 Study of Aln-AS1, an Investigational RNAi Therapeutic for the Treatment of Acute Hepatic Porphyria

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2318-2318 ◽  
Author(s):  
Eliane Sardh ◽  
Pauline Harper ◽  
Nabil Al-Tawil ◽  
Manisha Balwani ◽  
Karl Anderson ◽  
...  

Abstract Acute hepatic porphyria (AHP) is a family of rare metabolic disorders including acute intermittent porphyria (AIP), hereditary coproporphyria (HCP), and variegate porphyria (VP), caused by a deficiency in one of the eight enzymes required for heme biosynthesis in the liver. When ALA synthetase (ALAS1), the first and rate limiting step in the pathway, is induced by triggers such as exposure to certain drugs or fasting, the neurotoxic heme intermediates aminolevulinic acid (ALA) and porphobilinogen (PBG) can accumulate upstream of the deficient enzyme leading to acute and potentially life threatening neurovisceral attacks in AHP patients. RNA interference is a naturally occurring cellular mechanism mediated by small interfering RNA (siRNA) that allows for the inhibition of protein synthesis through the cleavage and degradation of a specific mRNA. ALN-AS1 is an investigational RNAi therapeutic that targets ALAS1 in order to decrease ALA and PBG levels and subsequent porphyria attacks. We are currently conducting a phase 1, multinational, randomized, placebo-controlled, study in 3 parts; Part A single ascending dose (SAD), Part B multiple ascending dose (MAD) and Part C multiple dose (MD) study to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics (i.e. changes in ALA, PBG and circulating ALAS1 mRNA levels) of subcutaneously administered ALN-AS1 in AIP patients. Part C also has exploratory analyses of clinical activity, including the impact of ALN-AS1 on porphyria attacks, acute healthcare visits, and heme treatment (ClinicalTrials.gov Identifier: NCT02452372). We previously presented interim SAD data demonstrating that ALN-AS1 was generally well tolerated with no serious adverse events (SAEs) or clinically significant laboratory abnormalities related to study drug, and no discontinuations due to AEs. Circulating ALAS1 mRNA levels had a mean (SEM) maximal reduction of 44% ± 8% relative to baseline (p ≤ 0.01 compared to placebo), with concomitant mean (SEM) maximal reductions in ALA and PBG of 77% ± 7% and 73% ± 6%, respectively (p= 0.03 and 0.06 compared to placebo, respectively) at the 0.35 mg/kg dose. In addition, changes in circulating ALAS1 mRNA were highly correlated with changes in urinary ALA and PBG (R2=0.82, p<10-15). We will now report interim progress and data from all 3 Parts of the phase 1 study including safety, pharmacodynamics, pharmacokinetics and exploratory clinical activity. Disclosures Sardh: Alnylam Pharmaceuticals: Consultancy. Harper:Alnylam Pharmaceuticals: Consultancy. Balwani:ICGG Gaucher Registry (Sanofi Genzyme sponsored): Membership on an entity's Board of Directors or advisory committees; Sanofi Genzyme: Honoraria, Speakers Bureau; Alnylam Pharmaceuticals: Consultancy. Anderson:Alnylam Pharmaceuticals: Consultancy. Bloomer:Alnylam Pharamceuticals: Consultancy. Bissel:Alnylam Pharmaceuticals: Consultancy. Desnick:Alnylam Pharmaceuticals: Consultancy. Bonkovsky:Alnylam Pharamceuticals: Consultancy. Penz:Alnylam Pharmaceuticals: Employment, Equity Ownership. Chan:Alnylam Pharmaceuticals: Employment, Equity Ownership. Soh:Alnylam Pharmaceuticals: Employment, Equity Ownership. Querbes:Alnylam Pharmaceuticals: Employment, Equity Ownership. Simon:Alnylam Pharmaceuticals: Employment, Equity Ownership. Rees:Alnylam Pharmaceuticals: Consultancy.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3891-3891 ◽  
Author(s):  
Anita Hill ◽  
Anna Gaya Valls ◽  
Morag Griffin ◽  
Talha Munir ◽  
Anna Borodovsky ◽  
...  

Abstract Background:Uncontrolled complement activation plays a pivotal role in a variety of disorders such as paroxysmal nocturnal hemoglobinuria (PNH). Challenges to be addressed with eculizumab therapy include inter-individual variation in clearance of eculizumab, economic burden, and improvements in the current biweekly intravenous infusion maintenance schedule. ALN-CC5 is a subcutaneous (SC) investigational RNA interference (RNAi) therapeutic targeting hepatic complement C5 (C5) synthesis. Previously presented data from our ongoing Phase 1/2 study showed that ALN-CC5 was generally well tolerated and exhibited a clamped C5 knockdown and complement activity inhibition in healthy volunteers (Hill et al. Haematologica 2016; 101, Suppl 1). The aim of this abstract is to report updated tolerability and clinical activity of ALN-CC5 in patients with PNH. Methods: A phase 1/2 single-ascending dose (Part A) and multiple-ascending dose study (Part B) of ALN-CC5 was conducted in healthy adult volunteers and in patients with PNH (Part C). In Part C, patients with PNH received weekly doses of 200 mg or 400 mg of ALN-CC5 for 2 to 16 weeks; ALN-CC5 is administered subcutaneously at a concentration of 200 mg/mL. The primary endpoints are safety and tolerability and secondary endpoints include: pharmacokinetics (PK), reduction of circulating C5 and complement activity, as measured by CAP/CCP Wieslab ELISA assays and sheep erythrocyte hemolysis assay, as well as reduction in LDH. Results: Part C included 6 patients with PNH (treatment naïve n=3; patients receiving eculizumab n=3), including 1 patient who was experiencing breakthrough hemolysis despite receiving 1200mg eculizumab q2wk. ALN-CC5 was generally well tolerated in patients with PNH after multiple doses with the majority of adverse events (AEs) being mild or moderate in severity. There were no serious AEs or discontinuations due to AEs. One patient, who was also taking eculizumab, cyclosporine and anabolic steroids as concomitant medications, experienced a transient asymptomatic grade 3 elevation of liver transaminases that was deemed possibly related to study drug. In eculizumab naïve patients (n=3), ALN-CC5 monotherapy achieved a mean maximum C5 knockdown of 98.2% ± 0.3%, residual C5 levels of 0.9 mcg/mL and a mean maximum CCP inhibition of 94.2 ± 1.7%. During treatment with ALN-CC5, maximum reduction in LDH was 37% and 50% in 2 patients who received 17 doses of ALN-CC5 but remained above the goal of less than 1.5 times the ULN. In the remaining eculizumab naïve patient, LDH lowering was not observed following 8 doses of ALN-CC5. After completion of ALN-CC5 dosing and in the setting of ongoing >95% ALN-CC5-mediated KD of serum C5, treatment naïve patients received a single 600 mg dose of eculizumab (labeled induction dose is 600 mg weekly x 4) for the treatment of residual hemolysis followed by close clinical monitoring. An exploratory analysis was conducted to understand the potential for ALN-CC5 to reduce eculizumab dose and frequency. All 3 patients achieved sustained lowering of LDH <1.5 ULN for out to 4 weeks. In patients entering the study on a stable dose of eculizumab (n=3), ALN-CC5 was also able to achieve a robust C5 KD (mean max 86.7% ± 5.6%) with residual complement activity of <2% as measured by CCP assay from day 21 onward. The addition of ALN-CC5 resulted in LDH lowering to within reference range by day 35 post treatment (maintained out to Day 112) in an inadequate response patient who entered the study on higher than labeled dose of eculizumab. As a result, this patient's subsequent dosing of eculizumab was lowered from 1200mg q2wk to 900mg q2wk. Updated safety, pharmacodynamics (PD) and clinical activity for all 6 patients with PNH in Part C of this ongoing Phase 1/2 study will be presented. Conclusion: ALN-CC5 was shown to be generally well tolerated in patients with PNH. The PD effects of ALN-CC5 were found to be durable, with clamped C5 knockdown and complement activity inhibition. Collectively, the data suggest that clamped inhibition of hepatic C5 synthesis may provide the foundation to potentially reduce the dose and frequency of eculizumab administration for patients with PNH, and to improve disease control in patients with inadequate response to eculizumab. Disclosures Hill: Alnylam Pharmaceuticals: Consultancy, Honoraria. Griffin:Alexion Pharmaceuticals: Honoraria, Other: Conference support. Munir:Alexion pharmaceuticals: Honoraria. Borodovsky:Alnylam Pharmaceuticals: Employment, Equity Ownership. Kawahata:Alnylam Pharmaceuticals: Employment, Equity Ownership. Mclean:Alnylam Pharmaceuticals: Employment, Equity Ownership. Shi:Alnylam Pharmaceuticals: Employment, Equity Ownership. Partisano:Alnylam: Employment, Equity Ownership. Kim:Alnylam Pharmaceuticals: Employment, Equity Ownership. Najafian:Alnylam Pharmaceuticals: Employment, Equity Ownership.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3920-3920 ◽  
Author(s):  
Marina Konopleva ◽  
Damian R Handisides ◽  
Mary Ann Richie ◽  
Juliana M Benito ◽  
Gautam Borthakur ◽  
...  

Abstract Background TH-302 is a 2-nitroimidazole prodrug of the DNA alkylator bromo-isophosphoramide mustard designed to be selectively activated in hypoxic conditions. Preclinical data in mice with acute lymphoblastic leukemia (ALL) have demonstrated marked expansion of hypoxia in areas of marrow leukemia infiltrates (Benito et al., PLoS One 2011). TH-302 also exhibited specific hypoxia-dependent cytotoxicity when tested against primary ALL and acute myelogenous leukemia (AML) samples in vitro (Benito et al., ASH 2012). Based on these findings, a phase 1 study of TH-302 was designed for advanced leukemias. Methods Eligible patients had ECOG ≤ 3, relapsed/refractory leukemias for which no standard therapy options were available, and acceptable hepato-renal function. A standard 3+3 dose-escalation design was used with 40% dose increments. TH-302 was administered IV over 30-60 min daily, either by 30 min-bolus administration or as a continuous infusion on days 1-5 of a 21-day cycle. The objectives were to determine the maximum tolerated dose (MTD) and pharmacokinetic (PK) profile of TH-302 with these schedules and to assess preliminary clinical activity of TH-302. Results A total of 49 patients with previously treated AML (n=39), ALL (n=9) or CML in blast phase (n=1) received TH-302 at bolus doses of 120 (n=4), 170 (n=4), 240 (n=3), 330 (n=3), 460 (n=20), 550 (n=4) mg/m2 or continuous doses of 330 (n=8) or 460 (n=3) mg/m2. Two of 3 evaluable patients treated with bolus TH-302 (550 mg/m2) experienced DLTs of grade 3 esophagitis; bolus administration MTD was established at 460 mg/m2. Two of 3 patients treated with continuous infusion of TH-302 (460 mg/m2) experienced DLTs of grade 3 mucositis or grade 3 hyperbilirubinemia; continuous administration MTD was established at 330 mg/m2. Thirteen patients received greater than 1 cycle. Generally, a significant rapid cytoreduction was evident early in the cycle, but was not maintained prior to initiation of the next cycle. Two AML patients who received 550 mg/m2 bolus TH-302 had complete resolution of leukemia cutis. One AML patient at 550 mg/m2 bolus TH-302 had a complete response with incomplete platelet recovery (CRp), and one AML patient at 440 mg/m2 bolus TH-302 had a CR. Conclusions In patients with advanced leukemias, MTDs were established for daily bolus infusion and 5-day continuous infusion of TH-302 at 460 mg/m2 and 330 mg/m2, respectively. Increased incidence of skin and mucosal toxicity were observed at higher dose levels with both administration schedules. Clinical activity of single-agent TH-302 has been noted with a few objective responses, but the majority of cytoreductions were transient. Disclosures: Konopleva: Threshold Pharmaceuticals: Research Funding. Handisides:Threshold Pharmaceuticals: Employment, Equity Ownership. Kroll:Threshold Pharmaceuticals: Employment, Equity Ownership. Kantarjian:Ariad: Research Funding; Novartis: Research Funding; Pfizer: Research Funding; BMS: Research Funding.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1959-1959 ◽  
Author(s):  
Jatin J Shah ◽  
Jeffrey A. Zonder ◽  
Adam Cohen ◽  
Donna Weber ◽  
Sheeba Thomas ◽  
...  

Abstract Abstract 1959 Background: Kinesin Spindle Protein (KSP) is required for cell cycle progression through mitosis. Inhibition of KSP induces mitotic arrest and cell death. ARRY-520 is a potent, selective KSP inhibitor. Cancers such as multiple myeloma (MM) which depend on the short-lived survival protein MCL-1 are highly sensitive to treatment with ARRY-520. ARRY-520 shows potent activity in preclinical MM models, providing a strong rationale for its clinical investigation in this disease. Methods: This Phase 1 study was designed to evaluate the safety and pharmacokinetics (PK) of ARRY-520 administered intravenously (IV) on Day 1 and Day 2 q 2 weeks without/with granulocyte-colony stimulating factor (G-CSF). Patients (pts) with relapsed/refractory (RR) MM with 2 prior lines of therapy (including both bortezomib and an immunomodulatory agent, unless ineligible for or refusing to receive this therapy) were eligible. Cohorts of at least 3 pts were enrolled in a classical 3 + 3 dose escalation design. Pts were treated for 2 cycles (4 weeks) to evaluate safety prior to dose escalation. Results: Twenty five pts have been treated to date, with a median age of 60 years (range 44–79) and a median of 5 prior regimens (range 2–16). All pts received prior bortezomib or carfilzomib, 21 pts received prior lenalidomide, 17 pts prior thalidomide, and 18 pts had a prior stem cell transplant. Pts received ARRY-520 without G-CSF at 1 mg/m2/day (n = 3), and at 1.25 mg/m2/day (n = 7, 6 evaluable). A dose-limiting toxicity (DLT) of Grade 4 neutropenia was observed at 1.25 mg/m2/day, and this was considered the maximum tolerated dose (MTD) without G-CSF. As neutropenia was the DLT, dose escalation with prophylactic G-CSF support was initiated, at doses of 1.5 mg/m2/day (n = 7, 6 evaluable), 2.0 mg/m2/day (n = 6) and 2.25 mg/m2/day (n = 2) with G-CSF. Both the 2.0 mg/m2/day and 2.25 mg/m2/day dose levels were determined to be non-tolerated, with DLTs of febrile neutropenia (FN) (2 pts at 2.0 mg/m2/day and both pts at 2.25 mg/m2/day) and Grade 3 mucositis (both pts at 2.25 mg/m2/day). One out of 6 evaluable pts at 1.5 mg/m2/day also developed a DLT of FN. In an attempt to optimize the Phase 2 dose, an intermediate dose level of 1.75 mg/m2/day with G-CSF is currently being evaluated. The most commonly reported treatment-related adverse events (AEs) include those observed with other KSP inhibitors, such as hematological AEs (thrombocytopenia, neutropenia, anemia, leukopenia), fatigue, mucositis and other gastro-intestinal AEs. Pts displayed linear PK, a low clearance and a moderate volume of distribution, with moderate-to-high inter-individual variability in PK parameters. The median terminal elimination half life is 65 hours. The preliminary efficacy signal as a single agent is encouraging with 2 partial responses (PR) observed to date per IMWG and EBMT criteria in a heavily pretreated population (23 evaluable pts). A bortezomib-refractory pt with 8 prior lines of therapy, including a tandem transplant, treated at 1 mg/m2/day of ARRY-520 obtained a PR after Cycle 6, with urine protein and kappa light chain levels continuing to decline over time. He remains on-study after 15 months of ARRY-520 treatment. A pt with 2 prior lines of therapy, including prior carfilzomib, has obtained a PR after Cycle 8 at 2 mg/m2/day of ARRY-520, and she is currently ongoing after 4.5 months on therapy. Fifteen pts had a best response of stable disease (SD), including 1 pt with a thus far unconfirmed minimal response, and 6 had progressive disease. A total of 10 pts (43%) achieved a PR or SD lasting > 12 weeks. Several additional pts have shown other evidence of clinical activity, with decrease in paraproteins, increase in hemoglobin levels and regression of plasmacytomas. The median number of cycles is 4 (range 1–28+). Treatment activity has not correlated with any baseline characteristics or disease parameters to date. Conclusions: : The selective KSP inhibitor ARRY-520 has been well tolerated, and shows promising signs of single agent clinical activity in heavily pretreated pts with RR MM. Prophylactic G-CSF has enabled higher doses to be tolerated. No cardiovascular or liver enzyme toxicity has been reported. Enrollment is ongoing at 1.75 mg/m2/day with G-CSF support, and a planned Phase 2 part of the study will be initiated as soon as the MTD is determined. Complete Phase 1 data will be disclosed at the time of the meeting. Disclosures: Shah: Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Millennium: Research Funding. Off Label Use: Revlimid (lenalidomide) in combination with dexamethasone is indicated for the treatment of multiple myeloma patients who have received at least one prior therapy. Zonder:Millennium: Consultancy, Myeloma and Amyloidosis Patient Day Symposium – Corporate support from multiple sponsors for a one-day educational event, Research Funding; Celgene:; Novartis:; Proteolix: . Weber:novartis-unpaid consultant: Consultancy; Merck- unpaid consultant: Consultancy; celgene- none for at least 2 years: Honoraria; millenium-none for 2 years: Honoraria; celgene, Millenium, Merck: Research Funding. Wang:Celgene: Research Funding; Onyx: Research Funding; Millenium: Research Funding; Novartis: Research Funding. Kaufman:Celgene: Consultancy, Honoraria, Research Funding; Millenium: Consultancy, Honoraria; Merck: Research Funding; Genzyme: Consultancy. Walker:Array Biopharma: Employment, Equity Ownership. Freeman:Array Biopharma: Employment, Equity Ownership. Rush:Array Biopharma: Employment, Equity Ownership. Ptaszynski:Array Biopharma: Consultancy. Lonial:Millennium, Celgene, Bristol-Myers Squibb, Novartis, Onyx: Advisory Board, Consultancy; Millennium, Celgene, Novartis, Onyx, Bristol-Myers Squibb: Research Funding.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 57-57 ◽  
Author(s):  
Jan A. Burger ◽  
Susan O'Brien ◽  
Nathan Fowler ◽  
Ranjana Advani ◽  
Jeff Porte Sharman ◽  
...  

Abstract Abstract 57 Introduction: Bruton's tyrosine kinase (Btk) is a downstream mediator of B-cell receptor (BCR) signaling and is not expressed in T-cells or NK-cells. As such, Btk represents an ideal therapeutic target for B-cell malignancies dependent upon BCR signaling. Chronic lymphocytic leukemia (CLL)/small lymphocytic leukemia (SLL) has been reported to have constitutively active BCR signaling. PCI-32765 is a potent, selective, irreversible and orally bioavailable small molecule inhibitor of Btk that has pre-clinical activity in B-cell malignancies (Proc Natl Acad Sci 2010;107(29):13075-80). PCI-32765 was therefore moved forward to a Phase 1 study in B-cell malignancies including patients (pts) with CLL/SLL. A subsequent CLL/SLL-specific Phase 1b study was initiated to further explore safety, pharmacokinetics (PK), pharmacodynamics (PD), and efficacy of PCI-32765. This report includes a composite summary of the CLL/SLL experience in both of these studies. Pts and Methods: Pts with CLL/SLL who had relapsed or refractory disease after >1 prior treatment regimens were eligible for treatment in each of the studies whereas the second Phase 1b study also included a cohort of elderly pts (aged ≥ 65 years) with CLL/SLL who required treatment and were “treatment-naive”. Responses were assessed by the investigator using the International Working Group CLL criteria (Hallek et al, Blood 2008 for pts with CLL) and the International Workshop to Standardize Response Criteria for Non-Hodgkin's Lymphomas (Cheson et al, J Clin Oncol 2007 for pts with SLL). Results: To date, 30 CLL/SLL patients (including 4 treatment-naive) have been enrolled across the 2 studies. Eighty-four percent of subjects are men with an overall median age of 68 (range 44–82) years. Of the subjects with prior therapy for CLL/SLL the median number of prior therapies is 3 (range 1–4). Treatment has been well-tolerated; Grade ≥ 3 toxicities have been infrequent (10/30 pts; 33%). Two study-drug related serious adverse events have been reported: 1 case of viral adenitis (Grade 3) and 1 case of viral infection (Grade 2). Two adverse events have led to discontinuation of study drug: a small bowel obstruction (Grade 3) and exacerbation of chronic obstructive disease (Grade 3); both events were reported as unrelated to study drug. No study-drug related deaths have reported. There has been no change in either NK cell or T cell counts. Target inhibition as measured by a probe of Btk drug occupancy showed inhibition of Btk at PCI-32765 exposure levels of ≥ 245 ng•h/mL. Of the 14 patients currently evaluable for response using the pre-defined criteria, the overall response rate is 64% (1 complete remission [CR], 8 partial remissions [PR], and 4 SD). Both studies are ongoing and open to enrollment. An update on response rate, response duration, safety, and PD information will be presented on enrolled patients based on a November 2010 database cut-off. Conclusion: PCI-32765 is a novel oral and selective “first-in-human” inhibitor of Btk that induces objective partial and complete responses in a substantial proportion of pts with CLL/SLL and has a favorable safety profile. These data support further studies of both monotherapy and also combination treatment with PCI-32765 in CLL/SLL. Disclosures: O'Brien: Pharmacyclics, Inc: Honoraria, PI grant. Fowler:Pharmacyclics: Consultancy, Research Funding. Advani:Pharmacyclics, Inc: Honoraria, PI grant. Sharman:Pharmacyclics, Inc: Honoraria, PI grant. Furman:Pharmacyclics, Inc: PI grant. Izumi:Pharmacyclics, Inc: Employment. Buggy:Pharmacyclics, Inc: Employment, Equity Ownership. Loury:Pharmacyclics: Employment, Equity Ownership. Hamdy:Pharmacyclics, Inc: Employment, Equity Ownership.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4496-4496
Author(s):  
Jennifer Brown ◽  
Ruth Plummer ◽  
Stephen Anthony ◽  
John Sarantopoulos ◽  
Filip De Vos ◽  
...  

Abstract Introduction: The PK profile of carfilzomib is well characterized in patients with multiple myeloma. However, during clinical development of carfilzomib, patients with moderate to severe hepatic impairment (HI) were excluded from initial clinical studies. To support carfilzomib dose recommendations for patients with baseline HI, this study evaluated PK and safety of carfilzomib in patients with varying degrees of HI and relapsed or progressive advanced malignancies. Methods: This open-label, single-arm, phase 1 study evaluated adult patients with normal (Norm) hepatic function or, mild, moderate (Mod), severe HI receiving carfilzomib infusion on days (D) 1-2, 8-9, 15 and 16 in 28-D cycles (C). Dose was escalated from 20 mg/m2 on C1 D1-D2 to 27 mg/m2 on D8 of C1 and if tolerated, further to 56 mg/m2 on D1 of C2. Norm hepatic function defined as bilirubin and aspartate aminotransferase (AST) levels </=upper limit of normal (ULN). HI defined as mild: bilirubin >1-1.5 x ULN, or AST >ULN but with bilirubin </=ULN; Mod: bilirubin >1.5-3 x ULN with any AST; or severe: bilirubin >3 x ULN and any AST. The primary objective was to assess the effect of HI on area under the curve (AUC) from time 0 to the last concentration measured (AUC0-last) and from time 0 extrapolated to infinity (AUC0-inf) of carfilzomib. Secondary objectives included evaluation of carfilzomib maximum plasma concentration (Cmax), time to maximum concentration (Tmax), clearance (CL), terminal half-life (T1/2), volume of distribution at steady state (Vss), mean residence time (MRT), and safety and tolerability, as well as PK parameters for carfilzomib's major metabolites. Plasma for analysis of PK parameters were collected on C1D16 for carfilzomib 27 mg/m2 and on C2D1 for the 56 mg/m2 dose. PK parameters were evaluated using a non-compartmental approach. The carfilzomib PK in HI patients was compared with Norm patients using summary statistics and analysis of variance. Due to enrollment challenges and lack of demonstrable efficacy with carfilzomib monotherapy, enrollment of severe HI patient (mostly advanced solid tumors) was discontinued. Results: 11 Norm, 17 Mild, 14 Mod, and 4 severe patients were enrolled; 61% male, mean age 62 years. Of these patients, 10 Norm, 14 Mild, 9 Mod, and 0 severe HI patients were PK evaluable. Following carfilzomib 27 and 56 mg/m2, considerable PK variability was seen within each of the treatment groups, with an overlapping exposure observed between groups (Table 1). Median Tmax ranged from 0.29 to 0.48 hour with peak concentrations of carfilzomib most often observed at 15 minutes after start or immediately before the end of infusion. Thereafter, concentrations of carfilzomib declined rapidly with a mean T1/2 of approximately 0.5 to 0.7 hour in all patient groups. A dose-dependent increase in mean AUC and Cmax of carfilzomib was observed between 27 mg/m2 and 56 mg/m2 in all 3 patient groups (Table 1); however, there was no consistent trend of increasing exposure (AUC0-last, AUC0-inf, and Cmax) with increasing severity of HI (Table 1 and 2). The mean AUC of the most abundant metabolite, PR-389/M14 was similar across all groups. A mean increase of approximately 60%-80% was observed for M15 and M16 AUC0-last, AUC0-inf and Cmax in patients with Mod HI vs Norm patients. These metabolites have no known biological activity. Median duration of exposure was 6 (Norm), 4.3 (Mild), 2.3 (Mod), and 0.8 (severe) wks. Thirty-five (76%) patients had grade >/=3 adverse events (AEs) including 15 patients with treatment-related grade >/=3 AEs. Grade >/=3 increased blood bilirubin (22%; Mod HI patients only), anemia (15%), fatigue (15%), and increased alanine aminotransferase (9%; Mod HI patients only) occurred in >3 patients. Conclusions: No marked differences in exposures (AUC and Cmax) were observed between Norm patients and mild/Mod HI patients following carfilzomib doses of 27 and 56 mg/m2.No consistent trend in carfilzomib exposure related to HI severity was seen. With the exception of the increased frequency of AEs consistent with hepatic function abnormalities, the observed AE profile in this study was consistent with the known safety profile of carfilzomib. HI did not appear to substantially increase severity of AEs; however, the number of patients was limited. Based on the results in this study, no carfilzomib dose adjustment appears to be warranted in patients with relapsed or progressive advanced malignancies and mild or Mod HI. Disclosures Anthony: Spectrum Pharmaceuticals: Speakers Bureau; Paradigm Diagnostics: Consultancy. De Vos:European Organization for Research and Treatment of Cancer: Consultancy, Membership on an entity's Board of Directors or advisory committees; Dutch Working Group Neuro-Oncology: Consultancy, Membership on an entity's Board of Directors or advisory committees; University Medical Center Utrecht: Employment. White:Amgen: Employment. Schupp:Amgen Inc.: Employment, Equity Ownership. Ou:Amgen: Employment, Equity Ownership.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4219-4219 ◽  
Author(s):  
Shaji K. Kumar ◽  
Ravi Vij ◽  
Jonathan L. Kaufman ◽  
Joseph R. Mikhael ◽  
Thierry Facon ◽  
...  

Abstract Background: The anti-apoptotic protein BCL-2 has been implicated in mediating the survival of multiple myeloma (MM) cells. Venetoclax is a potent, selective, orally bioavailable small-molecule BCL-2 inhibitor. Venetoclax induces cell death in MM cell lines in vitro and primary MM samples ex vivo. Certain genetic subtypes of MM cells are particularly sensitive to venetoclax, including t(11;14) cells, which express a high ratio of BCL2 to MCL1 (venetoclax resistance factor). The current Phase 1 study evaluates safety, efficacy, and pharmacokinetics (PK) in patients (pts) with relapsed/refractory MM. Methods: Primary objectives are to evaluate safety, PK, and recommended phase two dose; other objectives include assessing preliminary efficacy and the impact of chromosomal abnormalities. In dose-escalation (DE) cohorts, venetoclax was given orally daily at 300, 600, 900, or 1200 mg after a 2-week dose ramp-up (3+3 design). Patients in the safety expansion (SE) cohort received 1200 mg daily after ramp-up. All patients were monitored for tumor lysis syndrome (TLS). Results: As of June 17, 2015, 37 patients were enrolled in the study: 30 from DE cohorts and 7 from the SE. Median (range) age was 66 years; 19 (51%) were female. Fourteen were ISS stage I, 13 stage II, 8 stage III, 2 unknown. The median (range) number of prior lines of therapy was 6 (1-19). Thirty-two had prior bortezomib (20 refractory), 35 lenalidomide (18 refractory), and 26 had prior stem cell transplant. Fourteen patients had t(11;14), 4 had t(4;14), 5 had del 17p, and 17 had del 13q. Adverse events (AEs) in ≥20% of patients were nausea (49%), diarrhea (38%), vomiting (30%), anemia (27%), fatigue (24%). Grade 3/4 AEs (≥10%): thrombocytopenia (22%), anemia (19%), neutropenia (11 %). Serious AEs (≥2 patients): pyrexia (n=3), cough, malignant neoplasm progression, and sepsis (2 each); 2 (upper abdominal pain and anemia) were possibly related to venetoclax. Thirty (81%) patients have discontinued venetoclax: 24 due to PD, 3 for AEs (worsening shortness of breath, hypokalemia, and nausea), 2 withdrew consent, 1 due to death (brain hemorrhage following injury). Four deaths occurred (2 due to PD, 1 due to brain hemorrhage, 1 due to pneumopathy). Two of the 6 patients in the 600 mg cohort experienced DLTs of upper abdominal pain and nausea with abdominal pain. No patients met the criteria for laboratory or clinical TLS. Based on preliminary PK (n=21), the mean Cmax and AUC24 were ~dose-proportional at all studied doses (300, 600, 1200 mg) except 900 mg, and dose-normalized venetoclax exposure in MM was similar to that in CLL and NHL pts. Thirty-two of the 37 patients were evaluable for preliminary efficacy (Table). Two patients, both t(11;14), achieved a complete response (1 at 600 mg and 1 at 900 mg). Responses were first achieved at 1.8 and 1.1 months and were maintained for 9.7 and 9.0 months, respectively (900 mg pt remains in CR). Among the 16 patients receiving 1200 mg in the DE or SE cohort, 6 of whom had t(11;14), 5 achieved SD, 6 experienced PD, and 5 are not yet evaluable. Conclusions: Venetoclax monotherapy had a tolerable safety profile in heavily-pretreated relapsed/refractory MM, and no new safety signals were observed compared to other venetoclax studies. The study continues to enroll in the SE cohort at 1200 mg. Responses (including CR) and longer time on venetoclax were observed in t(11;14) patients. These early results suggest that venetoclax has single agent activity, most prominently in t(11;14) patients. Figure 1. Figure 1. Disclosures Kumar: Celgene: Research Funding; Millenium/Takeda: Research Funding; Onyx: Research Funding; AbbVie: Research Funding; Janssen: Research Funding; Sanofi: Research Funding; Celgene, Millenium, Sanofi, Skyline, BMS, Onyx, Noxxon,: Other: Consultant, no compensation,; Skyline, Noxxon: Honoraria. Off Label Use: Venetoclax is an investigational drug that is not yet approved in this indication.. Vij:Takeda, Onyx: Research Funding; Celgene, Onyx, Takeda, Novartis, BMS, Sanofi, Janssen, Merck: Consultancy. Kaufman:Janssen: Consultancy; Spectrum: Consultancy; Merck: Research Funding; Celgene: Consultancy; Onyx: Consultancy; Novartis: Consultancy; Novartis: Research Funding; Onyx: Research Funding. Mikhael:Sanofi: Research Funding; AbbVie: Research Funding; Celgene: Research Funding; Onyx: Research Funding. Moreau:Takeda: Other: Adboard; Janssen: Other: Adboard; Celgene: Other: Adboard; Novartis: Other: Adboard; Amgen: Other: Adboard. Alzate:AbbVie: Employment, Equity Ownership. Morris:AbbVie: Employment, Equity Ownership. Ross:AbbVie: Employment, Equity Ownership. Dunbar:AbbVie: Employment, Equity Ownership. Zhu:AbbVie: Employment, Equity Ownership. Maciag:AbbVie: Employment, Equity Ownership. Agarwal:AbbVie: Employment, Equity Ownership. Leverson:AbbVie: Employment, Equity Ownership. Enschede:AbbVie: Employment, Equity Ownership. Humerickhouse:AbbVie: Employment, Equity Ownership. Touzeau:AbbVie: Research Funding.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 56-56
Author(s):  
Richard R. Furman ◽  
Leslie Andritsos ◽  
Ian W. Flinn ◽  
Andres Forero-Torres ◽  
Kenneth A. Foon ◽  
...  

Abstract Abstract 56 Background: CD37 is a member of the tetraspanin superfamily of molecules which are implicated in diverse processes including cellular activation and proliferation, cell motility, and cell-cell adhesion. Studies in CD37-deficient mice suggest that CD37 is involved in the regulation of B-cell function, but is not required for B-cell development. CD37 is a heavily glycosylated cell surface protein expressed constitutively at high levels on normal and transformed B-cells across a wide range of maturational stages. TRU-016 is a novel humanized anti-CD37 SMIP™ protein, which is a single chain Fv-FC fusion protein. Pre-clinical studies have demonstrated that anti-CD37 SMIP protein mediates significantly greater direct killing of CLL cells than rituximab. TRU-016 also has greater Fc mediated cellular cytotoxicity of CLL cells than either alemtuzumab or rituximab. Methods: The objective of the Phase 1 study was to establish the maximum tolerated dose, overall safety and clinical activity of TRU-016 in patients with advanced CLL and SLL. Response was determined using the 1996 NCI working group criteria. Patients with relapsed/refractory CLL or SLL who had adequate organ function and platelets > 30,000/mm3 were eligible. Nine dose levels, ranging from 0.03 mg/kg to 20 mg/kg IV given once a week for 4 to 12 doses (weekly), were studied. A second schedule tested 3, 6 or 10 mg/kg on days 1, 3 and 5 the first week followed by 3 to 11 weekly doses (TIW). Dose escalation and de-escalation was based on NCI CTCAE toxicity grades. Results: 57 patients were treated with TRU-016. The median number of prior treatment regimens was 4; median number of prior anti-CD20 treatment regimens was 2 and in those with data available, 59% were refractory to their prior therapy for CLL. The median age was 66 years and 68.5% were Rai stage 3 or 4. Genomic data are available for 53 patients and 35 (66%) had high-risk genomic features [del(17p13.1), n=20 (38%), del(11q22.3), n=11 (21%), both=4 (8%)]. 19 patients reported serious adverse events (SAEs); the following events, regardless of causality, occurred in more than one patient: 3 febrile neutropenia, 3 pneumonia, 2 infusion reactions, 2 pyrexia, and 2 dyspnea. There was no apparent dose relationship to the SAEs. There were 3 dose limiting toxicities (DLTs); grade 4 neutropenia at 6 mg/kg, grade 4 thrombocytopenia (ITP) at 3 mg/kg TIW and grade 4 neutropenia at 15 mg/kg. There were no SAEs or DLTs at the highest dose of 20 mg/kg, so a maximum tolerated dose (MTD) was not reached. Mild to moderate (grade 1–2) infusion toxicity was observed on the day of infusion; the most common consisted of nausea (23%) and chills (21%). There were 2 serious infusion reactions; 1 grade 2 and the other grade 3. Both resolved with interruption of study drug infusion. Pharmacokinetic data demonstrate rapid clearance of TRU-016 in the lower dose cohorts. Accumulation was noted at the 3 mg/kg TIW and 6 mg/kg weekly and higher dose cohorts. Beginning with the 3 mg/kg TIW dose cohort, serum concentrations were usually maintained above 10 μ g/ml during treatment. 7 partial responses (PR) by investigator assessment of best response were reported, including 2 PRs in patients with del(17p13.1). For patients with 1 or 2 prior therapies, the ORR rate was 44% (7/16), all PRs, and the median reduction in peripheral lymphocytes was 92%. In patients with 3 or more prior therapies (n=41), no responses were obtained, although there was a median reduction in lymphocytes of 67% (n=21 with baseline lymphocytosis and end of treatment data available). Conclusions: TRU-016 treatment has a favorable safety profile and the MTD has not been reached. Partial responses with single agent TRU-016 have been observed in patients with 1–2 prior therapies including those with del(17p13.1). Given the demonstrated single-agent clinical activity of TRU-016 and synergistic or additive impact of TRU-016 with multiple agents in preclinical models, further dose escalation and combination drug trials of TRU-016 have been initiated. Disclosures: Forero-Torres: Trubion Pharmaceuticals: Research Funding. Singhal:Facet Biotech: Employment, Equity Ownership. Stromatt:Trubion Pharmaceuticals: Employment, Equity Ownership.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5555-5555
Author(s):  
Robert B Kleiman ◽  
Rajesh Chopra ◽  
de Haan Hans ◽  
Kristen Hege ◽  
Angela J James ◽  
...  

Abstract Introduction: A variety of chemically diverse drugs are known to alter cardiac repolarization and to produce sudden cardiac death. Current regulatory guidelines therefore require the characterization of a new drug's effect on cardiac repolarization, and in particular, the effects on the QTc interval in a thorough QT/QTc study (ICH E14 Guidance, 2005). QTc studies are usually performed with healthy volunteers but may be performed in the targeted patient (pt) population if the toxicity profile of a drug precludes its use in healthy individuals. Early-phase oncology dose-escalation studies generally include pharmacokinetic/pharmacodynamic (PK/PD) evaluations from relatively large pt populations, which could provide a robust data set for concurrent cardiac safety evaluation. We have combined these approaches and present the results from a first-in-human phase 1 study of CC-223, a potent and selective dual inhibitor of mechanistic target of rapamycin (mTOR) kinases. Patients and Methods: Adult (aged > 18 years) pts with histologically confirmed advanced non-Hodgkin lymphoma, multiple myeloma, or advanced, unresectable solid tumors who had progressed on (or were unable to tolerate) standard therapy or for whom standard therapy does not exist were eligible. The dose of CC-223, administered orally, ranged from 7.5 to 60.0 mg/day in 28-day continuous cycles. PK blood samples and corresponding triplicate electrocardiograms (ECG) were collected at 2 time points: predose and 1.5-3 hours after the dose in cycle 2. ECG analysis was performed on all pts who received ≥ 1 dose of CC-223 with baseline and on-treatment ECG results available. The primary cardiac assessment was the by time point change from baseline for cardiac interval duration measurements, including heart rate (HR), PR interval, QRS duration, and QTcF interval (QT interval corrected for HR using the Fridericia formula). Secondary analyses included a time-averaged central tendency analysis, analysis of morphology and measurement outliers, and PK/PD analysis of the relationship between the plasma concentration of CC-223 and its M1 metabolite vs QTcF change from baseline. Results: As of April 1, 2013, 158 pts met the requirements for inclusion in the ECG analysis, and of those, 149 were also included in the PK/PD analysis. The data revealed no effect of CC-223 on cardiac interval duration measurements. The by time point analyses of HR, PR, QRS, and QTcF demonstrated no clinically significant ECG effects of CC-223 during cycle 1 or subsequent cycles. The time-averaged central tendency analyses also demonstrated only small changes in cardiac interval duration. For QTcF, the time-averaged mean change from baseline was −18.5 to −1.0 ms in cycle 1 and −8.7 to 7.3 ms in subsequent cycles. There were few nonspecific ECG morphological findings and few measurement outliers. The PK/PD analysis showed no evidence of any significant exposure-effect relationship between CC-223 or the M1 metabolite and QTcF (Figure). The estimated QTcF change at the maximum concentration (Cmax) of 354 ng/mL for CC-223 was −0.2 ms, with an upper 1-sided 95% CI of 1.2 ms, and the estimated QTcF change at Cmaxof 1532 ng/mL for the M1 metabolite was 0.4 ms, with an upper 1-sided 95% CI of 1.8 ms. Adverse events (AEs) related to cardiac function included 2 reports of ventricular arrhythmias; neither event was serious, both resolved while the pts were on study, and neither were considered study drug related. Conclusions: The QTcF intervals and PK/PD relationships for CC-223 and its active metabolite revealed no significant effect on cardiac repolarization or other ECG parameters. This study used a robust data set in pts, avoided potential AE exposure in healthy volunteers, and had the additional benefit of fulfilling the QTc study requirement for new drugs by the Food and Drug Administration. As such, combining a phase 1 study with the required QTc analysis can be an improved, cost-effective approach for assessing cardiac safety during early drug development. Disclosures Kleiman: Celgene Corporation: Consultancy. Chopra:Celgene: Employment, Equity Ownership. Hans:Celgene Corporation: Consultancy. Hege:Celgene Corporation: Employment, Equity Ownership. James:Celgene Corporation: Equity Ownership. Wang:Celgene Corporation: Employment, Equity Ownership. Wu:Celgene Corporation: Employment, Equity Ownership. O'Mara:Celgene Corporation: Employment, Equity Ownership.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3178-3178
Author(s):  
Robert J. Laliberte ◽  
Paul F. Glidden ◽  
Brian L. Hamilton

Abstract Introduction: Despite recent advances in the diagnosis and management of myeloproliferative neoplasms (MPNs), treatment of essential thrombocythemia (ET) has remained largely unchanged since the introduction of anagrelide in the US during 1997. Anagrelide is indicated for the treatment of thrombocythemia, to reduce the elevated platelet count and risk of thrombosis, and to ameliorate symptoms including thrombohemorrhagic events. The primary pharmacological effect of anagrelide is inhibition of megakaryocyte hypermaturation leading to reduced platelet production. Anagrelide also inhibits cyclic AMP phosphodiesterase III (PDE3), and common drug related adverse events (AEs; e.g., headache, palpitations, fluid retention, nausea and diarrhea) are believed to be due to this mechanism. Although initiating treatment at low doses and slowly increasing the dose to reach a target decreased platelet count may mitigate AEs, over 20% of patients still withdraw from treatment due to poor tolerability. As the currently marketed anagrelide product is an immediate release (IR) formulation with peak plasma concentrations (Cmax) that may exceed that needed for platelet reduction and cause unwanted PDE3 inhibition and AEs, an alternate formulation that modifies this pharmacokinetic (PK) profile may improve patient tolerability, adherence and treatment outcomes. This has led to the development and study of a controlled-release (CR) formulation of anagrelide (GALE-401). Methods: 98 healthy adult subjects were enrolled among 5 Phase 1 clinical trials of anagrelide CR, including 12 placebo-control subjects and 86 subjects who received single or multiple doses ranging from 0.2 to 0.6 mg twice daily (b.i.d.) for up to 41 days. The trials included an open-label, single dose developmental study; two placebo-controlled multiple dose ranging studies; a food effect study; and a comparative crossover PK study vs. IR reference product. Safety parameters included routine laboratory, ECG, and clinical evaluations. PK was assessed by measurements of plasma anagrelide and its active metabolite using a validated HPLC-MS/MS method. Pharmacodynamic activity was assessed by daily platelet count determinations in the multiple dosing studies. Results: Single doses of anagrelide CR were well tolerated, and the only drug-related AE reported in 2 or more subjects was headache. In the b.i.d. dose-ranging studies, the frequency and severity of AEs were similar between anagrelide CR and placebo groups, with the exception of decreased platelet counts in subjects receiving anagrelide CR. All AEs were transient, mild or moderate in severity, and no severe or serious AEs were reported. Anagrelide CR demonstrated dose proportional PK characteristics. Following a single 0.5 mg dose in the fasted state, the mean time to maximum plasma concentration (Tmax) and terminal elimination half-life (t1/2) were 2.0±1.5 hrs and 10.4±9.3 hrs (mean ± SD), respectively; in contrast, Tmax and t1/2 following IR was 1.0±0.9 hrs and 1.4±0.2 hrs, respectively. Cmax and total plasma exposure (AUC0-inf) with anagrelide CR were reduced to 26% and 59% of IR, respectively. However, steady-state PK following 6 daily 0.5 mg b.i.d. doses of anagrelide CR or IR showed similar AUC0-inf values, while Cmax with anagrelide CR remained nearly unchanged (29%). Plasma exposure was higher when anagrelide CR was administered in the fed state, as demonstrated by the ratio of least-squares mean values for Cmax and AUC0-t, which were increased by 100% and 60%, respectively. The platelet lowering effect of anagrelide CR was evident in the 2 multiple dose ranging studies. In a placebo-controlled study of 0.2 to 0.6 mg b.i.d. dosing for up to 21 days, a dose-related decrease in platelet counts was observed, and the 0.6 mg cohort was halted early due to excessive platelet reductions. Anagrelide CR did not have a relevant impact on platelet function as assessed by template bleeding time. Figure 1 Figure 1. Conclusion: Anagrelide CR is a promising, novel formulation of anagrelide that exhibited the desired PK profile of a significantly reduced Cmax, while maintaining plasma exposure to induce platelet count reductions. The product was well tolerated with an AE profile that was not distinguishable from placebo. These data support the importance of an ongoing Phase 2 study in patients with MPN-related thrombocytosis, including ET. Disclosures Laliberte: Galena Biopharma, Inc.: Employment, Equity Ownership. Glidden:Galena Biopharma, Inc.: Consultancy, Equity Ownership, Patents & Royalties. Hamilton:Galena Biopharma, Inc.: Employment, Equity Ownership.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1310-1310 ◽  
Author(s):  
Bin Fan ◽  
Kha Le ◽  
Erika Manyak ◽  
Hua Liu ◽  
Malia Prahl ◽  
...  

Abstract INTRODUCTION: Somatic IDH1/2 mutations occur in multiple solid and hematologic tumors, including acute myeloid leukemia. Mutant IDH1/2 proteins have novel enzymatic activity, catalyzing the reduction of α-ketoglutarate to produce the oncometabolite, D-2-hydroxyglutarate (2-HG), which drives multiple oncogenic processes including impaired cellular differentiation. AG-120 is a first-in-class, oral, potent, reversible and selective inhibitor of the mutated IDH1 protein, and has been shown to lower 2-HG levels and restore cellular differentiation in IDH1-mutant primary human blast cells cultured ex vivo. AG-120 is currently being assessed in a first-in-human, phase 1 study enrolling patients with IDH1-mutant, advanced hematologic malignancies (NCT02074839). Our objective was to further assess the longitudinal pharmacokinetic/pharmacodynamic (PK/PD) data from the ongoing phase 1 study, including dose proportionality assessment of AG-120 exposure after single and multiple doses over time, and the influence of patient-intrinsic factors. METHODS: The AG-120 phase 1, open-label, dose-escalation and expansion study includes evaluation of safety, tolerability, maximum tolerated dose, PK/PD (including 2-HG levels) and clinical activity. Single-agent AG-120 is administered orally once (QD) or twice (BID) daily in continuous 28-day cycles. Patients included in this analysis received doses of 100 mg BID, 300 mg QD, 500 mg QD, 800 mg QD and 1200 mg QD (N=39). Blood, bone marrow and urine samples were collected at multiple time points for determination of PK/PD using qualified LC-MS/MS-based methods. Analyses were performed using WinNonLin®. RESULTS: AG-120 exposure exceeding the predicted efficacious exposure level was demonstrated at all dose levels following oral administration. Following both single (Day-3) and multiple (Cycle 1 Day 15 [C1D15] and Cycle 2 Day 1) dose administration, mean plasma exposures of AG-120 increased less than proportionally to dose, although plasma exposures were quite variable within dose levels. Preliminary PK data revealed a mean half-life of 119 ± 104 hr. Following multiple doses, most patients achieved steady state in Cycle 1, with ~2 to 3-fold accumulation in plasma observed. Furthermore, pre-dose AG-120 trough levels were maintained above the predicted efficacious exposure level throughout treatment (up to 11 cycles). Following a single dose of AG-120, plasma 2-HG levels gradually reduced over 3 days. After multiple doses, plasma 2-HG levels were reduced to levels seen in healthy volunteers (up to 99.7% inhibition) at all dose levels tested. Steady state 2-HG inhibition was reached at approximately C1D15 in most patients, and was maintained over the course of treatment (up to 11 cycles). Mean bone marrow 2-HG levels were also substantially reduced following multiple doses of AG-120 at all dose levels tested (up to 99.9% reduction compared with baseline). Plasma 2-HG levels showed a positive correlation with levels in bone marrow (r2=0.882, p<0.001) and urine (r2=0.528, p<0.001), with a stronger correlation being observed with the former tissue. There was no clear effect of patient-intrinsic factors such as body weight or body surface area on Cmaxor AUC within the ranges tested, although the current sample size is small. Population PK and PK/PD assessments will be conducted to confirm these findings. These analyses are based on data as of 1 May 2015; updated analyses will be presented. CONCLUSION: AG-120 exposure increased less than proportionally to dose following oral administration, with long half-life and maintenance of pre-dose levels above the predicted efficacious exposure, supporting QD dosing. In patients with IDH1 mutations, AG-120 inhibited plasma 2-HG to within levels found in healthy volunteers, and also inhibited 2-HG in bone marrow. Disclosures Fan: Agios Pharmaceuticals: Employment, Equity Ownership. Le:Agios Pharmceuticals: Employment, Equity Ownership. Manyak:Agios Pharmaceuticals: Employment. Liu:Agios: Employment. Prahl:Agios Pharmaceuticals: Employment, Equity Ownership. Bowden:Agios Pharmaceuticals: Employment. Biller:Agios Pharmaceuticals: Employment, Equity Ownership; Arbutus BioPharma (formerly Tekmira): Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Syros Pharmaceuticals: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Arvinas: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Denali: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Agresta:Agios: Employment, Equity Ownership. Yang:Agios Pharmaceuticals: Employment, Equity Ownership.


Sign in / Sign up

Export Citation Format

Share Document